51
|
Good Cop, Bad Cop: Defining the Roles of Δ40p53 in Cancer and Aging. Cancers (Basel) 2020; 12:cancers12061659. [PMID: 32585821 PMCID: PMC7352174 DOI: 10.3390/cancers12061659] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 01/10/2023] Open
Abstract
The tumour suppressor p53 is essential for maintaining DNA integrity, and plays a major role in cellular senescence and aging. Understanding the mechanisms that contribute to p53 dysfunction can uncover novel possibilities for improving cancer therapies and diagnosis, as well as cognitive decline associated with aging. In recent years, the complexity of p53 signalling has become increasingly apparent owing to the discovery of the p53 isoforms. These isoforms play important roles in regulating cell growth and turnover in response to different stressors, depending on the cellular context. In this review, we focus on Δ40p53, an N-terminally truncated p53 isoform. Δ40p53 can alter p53 target gene expression in both a positive and negative manner, modulating the biological outcome of p53 activation; it also functions independently of p53. Therefore, proper control of the Δ40p53: p53 ratio is essential for normal cell growth, aging, and responses to cancer therapy. Defining the contexts and the mechanisms by which Δ40p53 behaves as a "good cop or bad cop" is critical if we are to target this isoform therapeutically.
Collapse
|
52
|
Finnie PSB, Nader K. Amyloid Beta Secreted during Consolidation Prevents Memory Malleability. Curr Biol 2020; 30:1934-1940.e4. [PMID: 32243855 DOI: 10.1016/j.cub.2020.02.083] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/21/2020] [Accepted: 02/26/2020] [Indexed: 12/21/2022]
Abstract
Memory allows organisms to predict future events based on their prior sampling of the world. Rather than faithfully encoding each detail of related episodes, the brain is thought to incrementally construct probabilistic estimates of environmental statistics that are re-evaluated each time relevant events are encountered [1]. When faced with evidence that does not adequately fit mnemonic predictions, a process called reconsolidation can alter relevant memories to better recapitulate ongoing experience [2]. Conversely, when an ongoing event matches well-established predictions, reactivated memories tend to remain stable [3, 4]. In part, the brain may confer selective mnemonic stability by shifting cell-intrinsic mechanisms of plasticity induction [5], which could serve to constrain maladaptive updating of reliably predictive representations during anomalous events. Based on evidence of decreased cognitive flexibility and restricted synaptic plasticity in later life [6], we hypothesized that some prevalent age-associated neurobiological changes might in fact contribute to mnemonic stability [7]. Specifically, we predicted that amyloid beta (Aβ)-a peptide that often accumulates in the brains of individuals expressing senescent dementia [8-10]-is required for memory stabilization. Indeed, we observe elevated soluble Aβx-42 concentrations in the amygdala shortly after young adult rats form reconsolidation-resistant auditory fear memories. Suppressing secretases required for Aβ production immediately after learning prevents mnemonic stabilization, rendering these memories vulnerable to disruption by post-reactivation amnestic treatments. Thus, the seemingly pathogenic Aβ42 peptide may serve an adaptive physiological function during memory consolidation by engaging mechanisms that protect reliably predictive representations against subsequent modification.
Collapse
Affiliation(s)
- Peter S B Finnie
- Psychology Department, McGill University, 1205 Avenue Dr. Penfield, Montreal, QC H3A 1B1, Canada.
| | - Karim Nader
- Psychology Department, McGill University, 1205 Avenue Dr. Penfield, Montreal, QC H3A 1B1, Canada.
| |
Collapse
|
53
|
Longitudinal Cognitive Decline in a Novel Rodent Model of Cerebral Amyloid Angiopathy Type-1. Int J Mol Sci 2020; 21:ijms21072348. [PMID: 32231123 PMCID: PMC7177469 DOI: 10.3390/ijms21072348] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 11/17/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a small vessel disease characterized by β-amyloid (Aβ) accumulation in and around the cerebral blood vessels and capillaries and is highly comorbid with Alzheimer’s disease (AD). Familial forms of CAA result from mutations within the Aβ domain of the amyloid β precursor protein (AβPP). Numerous transgenic mouse models have been generated around expression of human AβPP mutants and used to study cerebral amyloid pathologies. While behavioral deficits have been observed in many AβPP transgenic mouse lines, relative to rats, mice are limited in behavioral expression within specific cognitive domains. Recently, we generated a novel rat model, rTg-DI, which expresses Dutch/Iowa familial CAA Aβ in brain, develops progressive and robust accumulation of cerebral microvascular fibrillar Aβ beginning at 3 months, and mimics many pathological features of the human disease. The novel rTg-DI model provides a unique opportunity to evaluate the severity and forms of cognitive deficits that develop over the emergence and progression of CAA pathology. Here, we present an in-depth, longitudinal study aimed to complete a comprehensive assessment detailing phenotypic disease expression through extensive and sophisticated operant testing. Cohorts of rTg-DI and wild-type (WT) rats underwent operant testing from 6 to 12 months of age. Non-operant behavior was assessed prior to operant training at 4 months and after completion of training at 12 months. By 6 months, rTg-DI animals demonstrated speed–accuracy tradeoffs that later manifested across multiple operant tasks. rTg-DI animals also demonstrated delayed reaction times beginning at 7 months. Although non-operant assessments at 4 and 12 months indicated comparable mobility and balance, rTg-DI showed evidence of slowed environmental interaction. Overall, this suggests a form of sensorimotor slowing is the likely core functional impairment in rTg-DI rats and reflects similar deficits observed in human CAA.
Collapse
|
54
|
Flores-Muñoz C, Gómez B, Mery E, Mujica P, Gajardo I, Córdova C, Lopez-Espíndola D, Durán-Aniotz C, Hetz C, Muñoz P, Gonzalez-Jamett AM, Ardiles ÁO. Acute Pannexin 1 Blockade Mitigates Early Synaptic Plasticity Defects in a Mouse Model of Alzheimer's Disease. Front Cell Neurosci 2020; 14:46. [PMID: 32265655 PMCID: PMC7103637 DOI: 10.3389/fncel.2020.00046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/18/2020] [Indexed: 12/11/2022] Open
Abstract
Synaptic loss induced by soluble oligomeric forms of the amyloid β peptide (sAβos) is one of the earliest events in Alzheimer’s disease (AD) and is thought to be the major cause of the cognitive deficits. These abnormalities rely on defects in synaptic plasticity, a series of events manifested as activity-dependent modifications in synaptic structure and function. It has been reported that pannexin 1 (Panx1), a nonselective channel implicated in cell communication and intracellular signaling, modulates the induction of excitatory synaptic plasticity under physiological contexts and contributes to neuronal death under inflammatory conditions. Here, we decided to study the involvement of Panx1 in functional and structural defects observed in excitatory synapses of the amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic (Tg) mice, an animal model of AD. We found an age-dependent increase in the Panx1 expression that correlates with increased Aβ levels in hippocampal tissue from Tg mice. Congruently, we also observed an exacerbated Panx1 activity upon basal conditions and in response to glutamate receptor activation. The acute inhibition of Panx1 activity with the drug probenecid (PBN) did not change neurodegenerative parameters such as amyloid deposition or astrogliosis, but it significantly reduced excitatory synaptic defects in the AD model by normalizing long-term potentiation (LTP) and depression and improving dendritic arborization and spine density in hippocampal neurons of the Tg mice. These results suggest a major contribution of Panx1 in the early mechanisms leading to the synaptopathy in AD. Indeed, PBN induced a reduction in the activation of p38 mitogen-activated protein kinase (MAPK), a kinase widely implicated in the early neurotoxic signaling in AD. Our data strongly suggest that an enhanced expression and activation of Panx1 channels contribute to the Aβ-induced cascades leading to synaptic dysfunction in AD.
Collapse
Affiliation(s)
- Carolina Flores-Muñoz
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Bárbara Gómez
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Elena Mery
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Paula Mujica
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Ivana Gajardo
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudio Córdova
- Laboratorio de Estructura y Función Celular, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Daniela Lopez-Espíndola
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigaciones Biomédicas, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudia Durán-Aniotz
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibáñez, Santiago de Chile, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Pablo Muñoz
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigaciones Biomédicas, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Arlek M Gonzalez-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Álvaro O Ardiles
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
| |
Collapse
|
55
|
Lloyd GM, Trejo-Lopez JA, Xia Y, McFarland KN, Lincoln SJ, Ertekin-Taner N, Giasson BI, Yachnis AT, Prokop S. Prominent amyloid plaque pathology and cerebral amyloid angiopathy in APP V717I (London) carrier - phenotypic variability in autosomal dominant Alzheimer's disease. Acta Neuropathol Commun 2020; 8:31. [PMID: 32164763 PMCID: PMC7068954 DOI: 10.1186/s40478-020-0891-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 01/30/2020] [Indexed: 12/14/2022] Open
Abstract
The discovery of mutations associated with familial forms of Alzheimer's disease (AD), has brought imperative insights into basic mechanisms of disease pathogenesis and progression and has allowed researchers to create animal models that assist in the elucidation of the molecular pathways and development of therapeutic interventions. Position 717 in the amyloid precursor protein (APP) is a hotspot for mutations associated with autosomal dominant AD (ADAD) and the valine to isoleucine amino acid substitution (V717I) at this position was among the first ADAD mutations identified, spearheading the formulation of the amyloid cascade hypothesis of AD pathogenesis. While this mutation is well described in multiple kindreds and has served as the basis for the generation of widely used animal models of disease, neuropathologic data on patients carrying this mutation are scarce. Here we present the detailed clinical and neuropathologic characterization of an APP V717I carrier, which reveals important novel insights into the phenotypic variability of ADAD cases. While age at onset, clinical presentation and widespread parenchymal beta-amyloid (Aβ) deposition are in line with previous reports, our case also shows widespread and severe cerebral amyloid angiopathy (CAA). This patient also presented with TDP-43 pathology in the hippocampus and amygdala, consistent with limbic predominant age-related TDP-43 proteinopathy (LATE). The APOE ε2/ε3 genotype may have been a major driver of the prominent vascular pathology seen in our case. These findings highlight the importance of neuropathologic examinations of genetically determined AD cases and demonstrate striking phenotypic variability in ADAD cases.
Collapse
Affiliation(s)
- Grace M Lloyd
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Jorge A Trejo-Lopez
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Pathology, University of Florida, Gainesville, FL, 32610, USA
| | - Yuxing Xia
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Karen N McFarland
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neurology, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32610, USA
| | - Sarah J Lincoln
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Benoit I Giasson
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Anthony T Yachnis
- Department of Pathology, University of Florida, Gainesville, FL, 32610, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.
- Department of Pathology, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
56
|
McKee CA, Lananna BV, Musiek ES. Circadian regulation of astrocyte function: implications for Alzheimer's disease. Cell Mol Life Sci 2020; 77:1049-1058. [PMID: 31578625 PMCID: PMC7098845 DOI: 10.1007/s00018-019-03314-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/26/2019] [Accepted: 09/19/2019] [Indexed: 12/13/2022]
Abstract
The circadian clock regulates rhythms in gene transcription that have a profound impact on cellular function, behavior, and disease. Circadian dysfunction is a symptom of aging and neurodegenerative diseases, and recent studies suggest a bidirectional relationship between impaired clock function and neurodegeneration. Glial cells possess functional circadian clocks which may serve to control glial responses to daily oscillations in brain activity, cellular stress, and metabolism. Astrocytes directly support brain function through synaptic interactions, neuronal metabolic support, neuroinflammatory regulation, and control of neurovascular coupling at blood and CSF barriers. Emerging evidence suggests that the astrocyte circadian clock may be involved in many of these processes, and that clock disruption could influence neurodegeneration by disrupting several aspects of astrocyte function. Here we review the literature surrounding circadian control of astrocyte function in health and disease, and discuss the potential implications of astrocyte clocks for neurodegeneration.
Collapse
Affiliation(s)
- Celia A McKee
- Department of Neurology, Washington University School of Medicine, Box 8111, 425 S. Euclid Ave, St. Louis, MO, 63105, USA
| | - Brian V Lananna
- Department of Neurology, Washington University School of Medicine, Box 8111, 425 S. Euclid Ave, St. Louis, MO, 63105, USA
| | - Erik S Musiek
- Department of Neurology, Washington University School of Medicine, Box 8111, 425 S. Euclid Ave, St. Louis, MO, 63105, USA.
| |
Collapse
|
57
|
Kosenko E, Tikhonova L, Alilova G, Urios A, Montoliu C. The Erythrocytic Hypothesis of Brain Energy Crisis in Sporadic Alzheimer Disease: Possible Consequences and Supporting Evidence. J Clin Med 2020; 9:jcm9010206. [PMID: 31940879 PMCID: PMC7019250 DOI: 10.3390/jcm9010206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/24/2022] Open
Abstract
Alzheimer’s disease (AD) is a fatal form of dementia of unknown etiology. Although amyloid plaque accumulation in the brain has been the subject of intensive research in disease pathogenesis and anti-amyloid drug development; the continued failures of the clinical trials suggest that amyloids are not a key cause of AD and new approaches to AD investigation and treatment are needed. We propose a new hypothesis of AD development based on metabolic abnormalities in circulating red blood cells (RBCs) that slow down oxygen release from RBCs into brain tissue which in turn leads to hypoxia-induced brain energy crisis; loss of neurons; and progressive atrophy preceding cognitive dysfunction. This review summarizes current evidence for the erythrocytic hypothesis of AD development and provides new insights into the causes of neurodegeneration offering an innovative way to diagnose and treat this systemic disease.
Collapse
Affiliation(s)
- Elena Kosenko
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, Pushchino 142290, Russia; (L.T.); (G.A.)
- Correspondence: or ; Tel.: +7-4967-73-91-68
| | - Lyudmila Tikhonova
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, Pushchino 142290, Russia; (L.T.); (G.A.)
| | - Gubidat Alilova
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, Pushchino 142290, Russia; (L.T.); (G.A.)
| | - Amparo Urios
- Hospital Clinico Research Foundation, INCLIVA Health Research Institute, 46010 Valencia, Spain; (A.U.); (C.M.)
| | - Carmina Montoliu
- Hospital Clinico Research Foundation, INCLIVA Health Research Institute, 46010 Valencia, Spain; (A.U.); (C.M.)
- Pathology Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
58
|
Dong Y, Stewart T, Bai L, Li X, Xu T, Iliff J, Shi M, Zheng D, Yuan L, Wei T, Yang X, Zhang J. Coniferaldehyde attenuates Alzheimer's pathology via activation of Nrf2 and its targets. Am J Cancer Res 2020; 10:179-200. [PMID: 31903114 PMCID: PMC6929631 DOI: 10.7150/thno.36722] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/02/2019] [Indexed: 01/22/2023] Open
Abstract
Background: Alzheimer's disease (AD) currently lacks a cure. Because substantial neuronal damage usually occurs before AD is advanced enough for diagnosis, the best hope for disease-modifying AD therapies likely relies on early intervention or even prevention, and targeting multiple pathways implicated in early AD pathogenesis rather than focusing exclusively on excessive production of β-amyloid (Aβ) species. Methods: Coniferaldehyde (CFA), a food flavoring and agonist of NF-E2-related factor 2 (Nrf2), was selected by multimodal in vitro screening, followed by investigation of several downstream effects potentially involved. Furthermore, in the APP/PS1 AD mouse model, the therapeutic effects of CFA (0.2 mmol kg-1d-1) were tested beginning at 3 months of age. Behavioral phenotypes related to learning and memory capacity, brain pathology and biochemistry, including Aβ transport, were assessed at different time intervals. Results: CFA promoted neuron viability and showed potent neuroprotective effects, especially on mitochondrial structure and functions. In addition, CFA greatly enhanced the brain clearance of Aβ in both free and extracellular vesicle (EV)-contained Aβ forms. In the APP/PS1 mouse model, CFA effectively abolished brain Aβ deposits and reduced the level of toxic soluble Aβ peptides, thus eliminating AD-like pathological changes in the hippocampus and cerebral cortex and preserving learning and memory capacity of the mice. Conclusion: The experimental evidence overall indicated that Nrf2 activation may contribute to the potent anti-AD effects of CFA. With an excellent safety profile, further clinical investigation of coniferaldehyde might bring hope for AD prevention/therapy.
Collapse
|
59
|
Zhou XL, Xu MB, Jin TY, Rong PQ, Zheng GQ, Lin Y. Preclinical Evidence and Possible Mechanisms of Extracts or Compounds from Cistanches for Alzheimer's Disease. Aging Dis 2019; 10:1075-1093. [PMID: 31595204 PMCID: PMC6764737 DOI: 10.14336/ad.2018.0815-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/15/2018] [Indexed: 12/19/2022] Open
Abstract
Currently, disease-modified strategies to prevent, halt or reverse the progress of Alzheimer's disease (AD) are still lacking. Previous studies indicated extracts or compounds from Cistanches (ECC) exert a potential neuroprotective effect against AD. Thus, we conducted a preclinical systematic review to assess preclinical evidence and possible mechanisms of ECC in experimental AD. A systematical searching strategy was carried out across seven databases from their inceptions to July 2018. Twenty studies with 1696 rats or mice were involved. Neurobehavioral function indices as primary outcome measures were established by the Morris water maze test (n = 11), step-down test (n = 10), electrical Y-maze test (n = 4), step-through test (n = 3), open field test (n = 2) and passage water maze test (n = 1). Compared with controls, the results of the meta-analysis showed ECC exerted a significant effect in decreasing the escape latency, error times and wrong reaction latency in both the training test and the retention test, and in increasing the exact time and the percentage of time in the platform-quadrant and the number of platform crossings (all P<0.01). In conclusion, ECC exert potential neuroprotective effects in experimental AD, mainly through mechanisms involving antioxidant stress and antiapoptosic effects, inhibiting Aβ deposition and tau protein hyperphosphorylation and promoting synapse protection. Thus, ECC could be a candidate for AD treatment and further clinical trials.
Collapse
Affiliation(s)
- Xiao-Li Zhou
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Meng-Bei Xu
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ting-Yu Jin
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pei-Qing Rong
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guo-Qing Zheng
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Lin
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
60
|
Sharma S, Chakravarthy H, Suresh G, Devanathan V. Adult Goat Retinal Neuronal Culture: Applications in Modeling Hyperglycemia. Front Neurosci 2019; 13:983. [PMID: 31607843 PMCID: PMC6756134 DOI: 10.3389/fnins.2019.00983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022] Open
Abstract
Culture of adult neurons of the central nervous system (CNS) can provide a unique model system to explore neurodegenerative diseases. The CNS includes neurons and glia of the brain, spinal cord and retina. Neurons in the retina have the advantage of being the most accessible cells of the CNS, and can serve as a reliable mirror to the brain. Typically, primary cultures utilize fetal rodent neurons, but very rarely adult neurons from larger mammals. Here, we cultured primary retinal neurons isolated from adult goat up to 10 days, and established an in vitro model of hyperglycemia for performing morphological and molecular characterization studies. Immunofluorescence staining revealed that approximately 30–40% of cultured cells expressed neuronal markers. Next, we examined the relative expression of cell adhesion molecules (CAMs) in adult goat brain and retina. We also studied the effect of different glucose concentrations and media composition on the growth and expression of CAMs in cultured retinal neurons. Hyperglycemia significantly enhances neurite outgrowth in adult retinal neurons in culture. Expression of CAMs such as Caspr1, Contactin1 and Prion is downregulated in the presence of high glucose. Hyperglycemia downregulates the expression of the transcription factor CCAAT/enhancer binding protein (C/EBP α), predicted to bind CAM gene promoters. Collectively, our study demonstrates that metabolic environment markedly affects transcriptional regulation of CAMs in adult retinal neurons in culture. The effect of hyperglycemia on CAM interactions, as well as related changes in intracellular signaling pathways in adult retinal neurons warrants further investigation.
Collapse
Affiliation(s)
- Sapana Sharma
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| | - Harshini Chakravarthy
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| | - Gowthaman Suresh
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| | - Vasudharani Devanathan
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| |
Collapse
|
61
|
Serum amyloid A1 is involved in amyloid plaque aggregation and memory decline in amyloid beta abundant condition. Transgenic Res 2019; 28:499-508. [PMID: 31407125 DOI: 10.1007/s11248-019-00166-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 08/02/2019] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, characterized by cognitive impairment, progressive neurodegeneration, and amyloid-β (Aβ) lesion. In the neuronal death and disease progression, inflammation is known to play an important role. Our previous study on acute-phase protein serum amyloid A1 (SAA1) overexpressed mice showed that the liver-derived SAA1 accumulated in the brain by crossing the brain blood barrier (BBB) and trigger the depressive-like behavior on mouse. Since SAA1 involved in immune responses in other diseases, we focused on the possibility that SAA1 may exacerbate the neuronal inflammation related to Alzheimer's disease. A APP/SAA overexpressed double transgenic mouse was generated using amyloid precursor protein overexpressed (APP)-c105 mice and SAA1 overexpressed mice to examine the function of SAA1 in Aβ abundant condition. Comparisons between APP and APP/SAA1 transgenic mice showed that SAA1 exacerbated amyloid aggregation and glial activation; which lead to the memory decline. Behavior tests also supported this result. Overall, overexpression of SAA1 intensified the neuronal inflammation in amyloid abundant condition and causes the greater memory decline compared to APP mice, which only expresses Aβ 1-42.
Collapse
|
62
|
Emmanuel IA, Olotu FA, Agoni C, Soliman MES. Deciphering the 'Elixir of Life': Dynamic Perspectives into the Allosteric Modulation of Mitochondrial ATP Synthase by J147, a Novel Drug in the Treatment of Alzheimer's Disease. Chem Biodivers 2019; 16:e1900085. [PMID: 30990952 DOI: 10.1002/cbdv.201900085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/16/2019] [Indexed: 12/31/2022]
Abstract
The discovery of J147 represented a significant milestone in the treatment of age-related disorders, which was further augmented by the recent identification of mitochondrial ATP synthase as the therapeutic target. However, the underlying molecular events associated with the modulatory activity of J147 have remained unresolved till date. Herein, we present, for the first time, a dynamical approach to investigate the allosteric regulation of mATP synthase by J147, using a reliable human αγβ protein model. The highlight of our findings is the existence of the J147-bound protein in distinct structural associations at different MD simulation periods coupled with concurrent open↔close transitions of the β catalytic and α allosteric (ATP5A) sites as defined by Cα distances (d), TriCα (Θ) and dihedral (φ) angular parameters. Firstly, there was an initial pairing of the αγ subunits away from the β subunit followed by the formation of the 'non-catalytic' αβ pair at a distance from the γ subunit. Interestingly, J147-induced structural arrangements were accompanied by the systematic transition of the β catalytic site from a closed to an open state, while there was a concurrent transition of the allosteric site from an open αE conformation to a closed state. Consequentially, J147 reduced the structural activity of the whole αγβ complex, while the unbound system exhibited high atomistic deviations and structural flexibility. Furthermore, J147 exhibited favorable binding at the allosteric site of mATP synthase with considerable electrostatic energy contributions from Gln215, Gly217, Thr219, Asp312, Asp313, Glu371 and Arg406. These findings provide details on the possible effects of J147 on mitochondrial bioenergetics, which could facilitate the structure-based design of novel small-molecule modulators of mATP synthase in the management of Alzheimer's disease and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Iwuchukwu A Emmanuel
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Fisayo A Olotu
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Clement Agoni
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| |
Collapse
|
63
|
Guo Q, Zheng X, Yang P, Pang X, Qian K, Wang P, Xu S, Sheng D, Wang L, Cao J, Lu W, Zhang Q, Jiang X. Small interfering RNA delivery to the neurons near the amyloid plaques for improved treatment of Alzheimer׳s disease. Acta Pharm Sin B 2019; 9:590-603. [PMID: 31193846 PMCID: PMC6543096 DOI: 10.1016/j.apsb.2018.12.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 09/29/2018] [Accepted: 11/15/2018] [Indexed: 02/02/2023] Open
Abstract
Gene therapy represents a promising treatment for the Alzheimer׳s disease (AD). However, gene delivery specific to brain lesions through systemic administration remains big challenge. In our previous work, we have developed an siRNA nanocomplex able to be specifically delivered to the amyloid plaques through surface modification with both CGN peptide for the blood–brain barrier (BBB) penetration and QSH peptide for β-amyloid binding. But, whether the as-designed nanocomplex could indeed improve the gene accumulation in the impaired neuron cells and ameliorate AD-associated symptoms remains further study. Herein, we prepared the nanocomplexes with an siRNA against β-site amyloid precursor protein-cleaving enzyme 1 (BACE1), the rate-limiting enzyme of Aβ production, as the therapeutic siRNA of AD. The nanocomplexes exhibited high distribution in the Aβ deposits-enriched hippocampus, especially in the neurons near the amyloid plaques after intravenous administration. In APP/PS1 transgenic mice, the nanocomplexes down-regulated BACE1 in both mRNA and protein levels, as well as Aβ and amyloid plaques to the level of wild-type mice. Moreover, the nanocomplexes significantly increased the level of synaptophysin and rescued memory loss of the AD transgenic mice without hematological or histological toxicity. Taken together, this work presented direct evidences that the design of precise gene delivery to the AD lesions markedly improves the therapeutic outcome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Wei Lu
- Corresponding authors. Tel.: +86 21 519980068; fax: +86 21 51980067.
| | - Qizhi Zhang
- Corresponding authors. Tel.: +86 21 519980068; fax: +86 21 51980067.
| | | |
Collapse
|
64
|
Lehmann S, Paquet C, Malaplate-Armand C, Magnin E, Schraen S, Quillard-Muraine M, Bousiges O, Delaby C, Dumurgier J, Hugon J, Sablonnière B, Blanc F, Wallon D, Gabelle A, Laplanche JL, Bouaziz-Amar E, Peoc'h K. Diagnosis associated with Tau higher than 1200 pg/mL: Insights from the clinical and laboratory practice. Clin Chim Acta 2019; 495:451-456. [PMID: 31051163 DOI: 10.1016/j.cca.2019.04.081] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 01/25/2023]
Abstract
CONTEXT Cerebrospinal fluid (CSF) biomarkers are valuable tools for the diagnosis of neurological diseases. We aimed to investigate within a retrospective multicentric study the final diagnosis associated with very high CSF Tau levels and to identify patterns of biomarkers that would differentiate them in clinical practice, to help clinical biologists into physicians' counseling. PATIENTS AND METHODS Within the national multicentric network ePLM, we included 1743 patients from January 1, 2008, to December 31, 2013, with CSF biomarkers assayed by the same Innotest assays (protein Tau, phospho-Tau [pTau], and Aβ 1-42). We identified 205 patients with protein Tau concentration higher than 1200 pg/mL and final diagnosis. RESULTS Among those patients, 105 (51.2%) were suffering from Alzheimer's disease, 37 (18%) from sporadic Creuztfeldt-Jakob disease, and 63 (30.7%) from other neurological diseases including paraneoplastic/ central nervous system tumor, frontotemporal dementia, other diagnoses, amyloid angiopathy, Lewy body dementia, and infections of the central nervous system. Phospho-Tau, Aβ1-42 and Aβ1-42/pTau values differed significantly between the three groups of patients (p < .001). An Aβ1-42/pTau ratio between 4.7 and 9.7 was suggestive of other neurological diseases (threshold in AD: 8.3). CSF 14-3-3 was useful to discriminate Alzheimer's disease from Creuztfeldt-Jakob disease in case of Aβ1-42 concentrations <550 pg/mL or pTau>60 pg/mL. CONCLUSION This work emphasizes the interest of a well-thought-out interpretation of CSF biomarkers in neurological diseases, particularly in the case of high Tau protein concentrations in the CSF.
Collapse
Affiliation(s)
- S Lehmann
- CHU de Montpellier and Université de Montpellier, IRMB, CRB, Laboratoire de Biochimie et Protéomique Clinique, 80 Avenue Augustin Fliche, 34295 Montpellier, France
| | - C Paquet
- Centre de Neurologie Cognitive, Groupe Hospitalier Saint-Louis Lariboisière Fernand-Widal APHP, INSERM U942, Université Paris Diderot, France
| | - C Malaplate-Armand
- Laboratoire de Biochimie et Biologie Moléculaire, UF Oncologie - Endocrinologie - Neurobiologie, Hôpital Central, Centre Hospitalier Universitaire, Nancy, France
| | - E Magnin
- Centre Mémoire Ressources Recherche Besançon Franche-Comté, Departement of Neurology, CHU Besançon, Besançon, France
| | - S Schraen
- Univ.Lille, Inserm, CHU-Lille, UMR-S1172 and Neurobiology Unit, Centre de Biologie-Pathologie, Lille, France
| | | | - O Bousiges
- Laboratoire de Biochimie et de Biologie Moléculaire, Hôpital de Hautepierre, Hôpitaux Universitaire de Strasbourg, Strasbourg, France; Laboratoire de Neurosciences cognitives et Adaptatives (LNCA), UMR7364 Unistra/CNRS, Strasbourg, France
| | - C Delaby
- CHU de Montpellier and Université de Montpellier, IRMB, CRB, Laboratoire de Biochimie et Protéomique Clinique, 80 Avenue Augustin Fliche, 34295 Montpellier, France
| | - J Dumurgier
- Centre de Neurologie Cognitive, Groupe Hospitalier Saint-Louis Lariboisière Fernand-Widal APHP, INSERM U942, Université Paris Diderot, France
| | - J Hugon
- Centre de Neurologie Cognitive, Groupe Hospitalier Saint-Louis Lariboisière Fernand-Widal APHP, INSERM U942, Université Paris Diderot, France
| | - B Sablonnière
- Centre Mémoire Ressources Recherche Besançon Franche-Comté, Departement of Neurology, CHU Besançon, Besançon, France
| | - F Blanc
- 2ICube laboratory and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS-Neurocrypto, University of Strasbourg and CNRS, Strasbourg, France
| | - D Wallon
- Inserm U1079, University of Rouen, Department of Neurology, France
| | - A Gabelle
- Centre Mémoire Ressources Recherche, CHU de Montpellier, Hôpital Gui de Chauliac, Montpellier, Université Montpellier, Montpellier, France
| | - J L Laplanche
- Service de Biochimie et Biologie moléculaire, GH Saint-Louis-Lariboisière-Fernand Widal, APHP, Paris, France
| | - E Bouaziz-Amar
- Service de Biochimie et Biologie moléculaire, GH Saint-Louis-Lariboisière-Fernand Widal, APHP, Paris, France
| | - K Peoc'h
- Service de Biochimie et Biologie moléculaire, GH Saint-Louis-Lariboisière-Fernand Widal, APHP, Paris, France; APHP, HUPNVS, Hôpital Beaujon, Biochimie clinique, Clichy, France; Université Paris Diderot, France.
| | | |
Collapse
|
65
|
Reverting Metabolic Dysfunction in Cortex and Cerebellum of APP/PS1 Mice, a Model for Alzheimer's Disease by Pioglitazone, a Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) Agonist. Mol Neurobiol 2019; 56:7267-7283. [PMID: 31016475 DOI: 10.1007/s12035-019-1586-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/26/2019] [Indexed: 12/16/2022]
Abstract
Identification of molecular mechanisms underlying early-stage Alzheimer's disease (AD) is important for the development of new therapies against and diagnosis of AD. In this study, gas chromatography time-of-flight mass spectrometry (GC-TOF-MS)-based metabolomics approach was employed to investigate the metabolic profiles in plasma and brain tissues harvested from 5-month-old APP/PS1 transgenic mice and their wildtype counterparts. Since different brain regions were expected to have their own distinct metabolic signals, four different brain regions, namely cortex, hippocampus, midbrain and cerebellum tissues, were dissected and had their metabolic profiles studied separately. Biochemical assays were also performed on plasma and brain cortex tissue of transgenic mice and wildtype mice, with a focus on mitochondrial health. Amyloid precursor protein and amyloid-β levels in plasma, brain cortex tissue and mitochondria fractions isolated from brain cortex were measured to assess the amyloid pathology. Our findings include the observation of extensive metabolic alterations in cortex and cerebellum of APP/PS1 mice, but not in their hippocampus, midbrain and plasma. The major pathways affected in cortex and cerebellum of APP/PS1 mice were closely related to impaired energy metabolism and perturbation of amino acid metabolism in these mice. APP/PS1 mice also exhibited higher amyloid-β40 and amyloid-β42 in their cortex, accumulation of mitochondria APP in their cortex, and presented an altered oxidative state in their brain. Treatment with the peroxisome proliferator-activated receptor gamma (PPARγ) agonist pioglitazone (PIO) successfully restored the energy metabolism, lowered amyloid-β levels and afforded the APP/PS1 mice a better antioxidative capacity in their cortex.
Collapse
|
66
|
Cao K, Xiang J, Dong YT, Xu Y, Li Y, Song H, Zeng XX, Ran LY, Hong W, Guan ZZ. Exposure to fluoride aggravates the impairment in learning and memory and neuropathological lesions in mice carrying the APP/PS1 double-transgenic mutation. Alzheimers Res Ther 2019; 11:35. [PMID: 31010414 PMCID: PMC6477877 DOI: 10.1186/s13195-019-0490-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/04/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is responsible for 60-70% of all cases of dementia. On the other hand, the tap water consumed by hundreds of millions of people has been fluoridated to prevent tooth decay. However, little is known about the influence of fluoride on the expression of APP and subsequent changes in learning and memory and neuropathological injury. Our aim here was to determine whether exposure to fluoride aggravates the neuropathological lesions in mice carrying the amyloid precursor protein (APP)/presenilin1 (PS1) double mutation. METHODS These transgenic or wide-type (WT) mice received 0.3 ml of a solution of fluoride (0.1 or 1 mg/ml, prepared with NaF) by intragastric administration once each day for 12 weeks. The learning and memory of these animals were assessed with the Morris water maze test. Senile plaques, ionized calcium binding adaptor molecule 1 (Iba-1), and complement component 3 (C3) expression were semi-quantified by immunohistochemical staining; the level of Aβ42 was detected by Aβ42 enzyme-linked immunosorbent assays (ELISAs); the levels of synaptic proteins and enzymes that cleave APP determined by Western blotting; and the malondialdehyde (MDA) content and activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) measured by biochemical procedures. RESULTS The untreated APP mice exhibited a decline in learning and memory after 12 weeks of fluoride treatment, whereas treatment of these some animals with low or high levels of fluoride led to such declines after only 4 or 8 weeks, respectively. Exposure of APP mice to fluoride elevated the number of senile plaques and level of Aβ42, Iba-1, and BACE1, while reducing the level of ADAM10 in their brains. The lower levels of synaptic proteins and enhanced oxidative stress detected in the hippocampus of APP mice were aggravated to fluoride. CONCLUSIONS These findings indicate that exposure to fluoride, even at lower concentration, can aggravate the deficit in learning and memory and neuropathological lesions of the mice that express the high level of APP.
Collapse
Affiliation(s)
- Kun Cao
- Department of Pathology at the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
| | - Jie Xiang
- Department of Pathology at the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
| | - Yang-Ting Dong
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| | - Yi Xu
- Department of Pathology at the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
| | - Yi Li
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| | - Hui Song
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| | - Xiao-Xiao Zeng
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| | - Long-Yan Ran
- Department of Pathology at the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
| | - Wei Hong
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| | - Zhi-Zhong Guan
- Department of Pathology at the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| |
Collapse
|
67
|
Macrophagic scavenging of Aβ. Clin Neuropathol 2019; 38:48-50. [PMID: 30704554 PMCID: PMC6657423 DOI: 10.5414/np301175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2019] [Indexed: 11/19/2022] Open
Abstract
No abstract available.
Collapse
|
68
|
Belfiore R, Rodin A, Ferreira E, Velazquez R, Branca C, Caccamo A, Oddo S. Temporal and regional progression of Alzheimer's disease-like pathology in 3xTg-AD mice. Aging Cell 2019; 18:e12873. [PMID: 30488653 PMCID: PMC6351836 DOI: 10.1111/acel.12873] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/16/2018] [Accepted: 10/14/2018] [Indexed: 12/30/2022] Open
Abstract
Accumulation of amyloid-β (Aβ) and fibrillary tangles, as well as neuroinflammation and memory loss, are hallmarks of Alzheimer's disease (AD). After almost 15 years from their generation, 3xTg-AD mice are still one of the most used transgenic models of AD. Converging evidence indicates that the phenotype of 3xTg-AD mice has shifted over the years and contradicting reports about onset of pathology or cognitive deficits are apparent in the literature. Here, we assessed Aβ and tau load, neuroinflammation, and cognitive changes in 2-, 6-, 12-, and 20-month-old female 3xTg-AD and nontransgenic (NonTg) mice. We found that ~80% of the mice analyzed had Aβ plaques in the caudal hippocampus at 6 months of age, while 100% of them had Aβ plaques in the hippocampus at 12 months of age. Cortical Aβ plaques were first detected at 12 months of age, including in the entorhinal cortex. Phosphorylated Tau at Ser202/Thr205 and Ser422 was apparent in the hippocampus of 100% of 6-month-old mice, while only 50% of mice showed tau phosphorylation at Thr212/Ser214 at this age. Neuroinflammation was first evident in 6-month-old mice and increased as a function of age. These neuropathological changes were clearly associated with progressive cognitive decline, which was first apparent at 6 months of age and became significantly worse as the mice aged. These data indicate a consistent and predictable progression of the AD-like pathology in female 3xTg-AD mice, and will facilitate the design of future studies using these mice.
Collapse
Affiliation(s)
- Ramona Belfiore
- The Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, Arizona
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Alexis Rodin
- The Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Eric Ferreira
- The Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Ramon Velazquez
- The Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Caterina Branca
- The Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Antonella Caccamo
- The Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Salvatore Oddo
- The Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| |
Collapse
|
69
|
Medeiros ADM, Silva RH. Sex Differences in Alzheimer’s Disease: Where Do We Stand? J Alzheimers Dis 2019; 67:35-60. [DOI: 10.3233/jad-180213] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- André de Macêdo Medeiros
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Center of Health and Biological Sciences, Universidade Federal Rural do Semiárido, Mossoró, Brazil
| | - Regina Helena Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
70
|
Bouter C, Henniges P, Franke TN, Irwin C, Sahlmann CO, Sichler ME, Beindorff N, Bayer TA, Bouter Y. 18F-FDG-PET Detects Drastic Changes in Brain Metabolism in the Tg4-42 Model of Alzheimer's Disease. Front Aging Neurosci 2019; 10:425. [PMID: 30670962 PMCID: PMC6333025 DOI: 10.3389/fnagi.2018.00425] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/07/2018] [Indexed: 12/19/2022] Open
Abstract
The evaluation of new therapeutic strategies in Alzheimer’s disease (AD) relies heavily on in vivo imaging and suitable animal models that mimic the pathological changes seen in patients. 18F-Fluorodeoxyglucose (18F-FDG)-positron-emission tomography (PET) is a well-established non-invasive imaging tool for monitoring changes in cerebral brain glucose metabolism in vivo. 18F-FDG-PET is used as a functional biomarker for AD as patients show an early and progressive reduction of cerebral glucose metabolism. However, earlier studies in preclinical models of AD showed conflicting results. The aim of this study was the evaluation of cerebral glucose metabolism in the Tg4–42 mouse model of AD using 18F-FDG-PET/magnetic resonance imaging (MRI). Tg4–42 mice show an age-dependent reduction in glucose metabolism together with severe neuron loss and memory deficits. Similar to AD patients early decrease in 18F-FDG uptake was already detected in young (3 months) Tg4–42 mice. The altered glucose metabolism coupled with age- and disease related cognitive decline of Tg4–42 mice make it a well-suited model for preclinical testing of AD-relevant therapeutics.
Collapse
Affiliation(s)
- Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Philipp Henniges
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Timon N Franke
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Caroline Irwin
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Carsten Oliver Sahlmann
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Marius E Sichler
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité-University Medicine Berlin, Berlin, Germany
| | - Thomas A Bayer
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
71
|
Tan C, Dong Y, Wang J, Yang X. Vanadyl acetylacetonate attenuates Aβ pathogenesis in APP/PS1 transgenic mice depending on the intervention stage. NEW J CHEM 2019. [DOI: 10.1039/c9nj00820a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
VAC treatment caused different Grp75 responses before and after Aβ plaque formation.
Collapse
Affiliation(s)
- Chang Tan
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| | - Yaqiong Dong
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| | - Jing Wang
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| | - Xiaoda Yang
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| |
Collapse
|
72
|
Bieri G, Lucin KM, O'Brien CE, Zhang H, Villeda SA, Wyss-Coray T. Proteolytic cleavage of Beclin 1 exacerbates neurodegeneration. Mol Neurodegener 2018; 13:68. [PMID: 30594228 PMCID: PMC6310967 DOI: 10.1186/s13024-018-0302-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022] Open
Abstract
Background Neuronal cell loss contributes to the pathology of acute and chronic neurodegenerative diseases, including Alzheimer’s disease (AD). It remains crucial to identify molecular mechanisms sensitizing neurons to various insults and cell death. To date, the multifunctional, autophagy-related protein Beclin 1 has been shown to be both necessary and sufficient for neuronal integrity in neurodegenerative models associated with protein aggregation. Interestingly, besides its role in cellular homeostasis, Beclin 1 has also been ascribed a role in apoptosis. This makes it critical to elucidate whether Beclin 1 regulates neuronal death and survival across neurodegenerative conditions independent of protein clearance. Here, we provide experimental evidence for a direct functional link between proteolytic cleavage of Beclin 1 and apoptotic neuronal cell loss in two independent models of neurodegeneration in vivo. Methods Proteolytic cleavage of Beclin 1 was characterized in lysates of human AD brain samples. We developed viral tools allowing for the selective neuronal expression of the various Beclin 1 forms, including Beclin 1 cleavage products as well as a cleavage-resistant form. The effect of these Beclin 1 forms on survival and integrity of neurons was examined in models of acute and chronic neurodegeneration in vitro and in vivo. Markers of neuronal integrity, neurodegeneration and inflammation were further assessed in a Kainic acid-based mouse model of acute excitotoxic neurodegeneration and in a hAPP-transgenic mouse model of AD following perturbation of Beclin 1 in the susceptible CA1 region of the hippocampus. Results We find a significant increase in caspase-mediated Beclin 1 cleavage fragments in brain lysates of human AD patients and mimic this phenotype in vivo using both an excitotoxic and hAPP-transgenic mouse model of neurodegeneration. Surprisingly, overexpression of the C-terminal cleavage-fragment exacerbated neurodegeneration in two distinct models of degeneration. Local inhibition of caspase activity ameliorated neurodegeneration after excitotoxic insult and prevented Beclin 1 cleavage. Furthermore, overexpression of a cleavage-resistant form of Beclin 1 in hippocampal neurons conferred neuroprotection against excitotoxic and Amyloid beta-associated insults in vivo. Conclusions Together, these findings indicate that the cleavage state of Beclin 1 determines its functional involvement in both neurodegeneration and neuroprotection. Hence, manipulating the cleavage state of Beclin 1 may represent a therapeutic strategy for preventing neuronal cell loss across multiple forms of neurodegeneration. Electronic supplementary material The online version of this article (10.1186/s13024-018-0302-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gregor Bieri
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Neurosciences PhD Program, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kurt M Lucin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Caitlin E O'Brien
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hui Zhang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Saul A Villeda
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA. .,Center for Tissue Regeneration, Repair and Restoration, VA Palo Alto Health Care System, 3801 Miranda Avenue, 154W, Palo Alto, CA, 94304, USA.
| |
Collapse
|
73
|
Modeling amyloid beta and tau pathology in human cerebral organoids. Mol Psychiatry 2018; 23:2363-2374. [PMID: 30171212 PMCID: PMC6594704 DOI: 10.1038/s41380-018-0229-8] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/24/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022]
Abstract
The typical abnormalities observed in the brain of Alzheimer's disease (AD) patients include synaptic alterations, neuronal death, brain inflammation, and the accumulation of protein aggregates in the form of amyloid plaques and neurofibrillary tangles. Despite the development of many animal and in vitro models for AD, there is a lack of an experimental approach that fully recapitulates essential aspects of the disease in human cells. Here, we report the generation of a new model to study AD, consisting of cerebral organoids (COs) produced from human-induced pluripotent stem cells (iPSCs). Under our experimental conditions, COs grow to form three-dimensional (3D) structures containing neural areas with cortical-like organization. Analysis of COs by histological and biochemical methods revealed that organoids produced from iPSCs derived from patients affected by familial AD or Down syndrome (DS) spontaneously develop over time pathological features of AD, including accumulation of structures highly reminiscent to amyloid plaques and neurofibrillary tangles. These pathological abnormalities were not observed in COs generated from various controls, including human iPSCs from healthy individuals, human iPSCs from patients affected by Creutzfeldt-Jakob disease, mouse embryonic stem cells (ESCs), or mouse iPSCs. These findings enable modeling genetic AD in a human cellular context in a 3D cortical-like tissue developed in vitro from patient-specific stem cells. This system provides a more relevant disease model compared to pre-existing methods and offers a new platform for discovery of novel targets and screening of drugs for therapeutic intervention.
Collapse
|
74
|
Suh M, Lee DS. Brain Theranostics and Radiotheranostics: Exosomes and Graphenes In Vivo as Novel Brain Theranostics. Nucl Med Mol Imaging 2018; 52:407-419. [PMID: 30538772 PMCID: PMC6261865 DOI: 10.1007/s13139-018-0550-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/10/2018] [Accepted: 10/05/2018] [Indexed: 12/17/2022] Open
Abstract
Brain disease is one of the greatest threats to public health. Brain theranostics is recently taking shape, indicating the treatments of stroke, inflammatory brain disorders, psychiatric diseases, neurodevelopmental disease, and neurodegenerative disease. However, several factors, such as lack of endophenotype classification, blood-brain barrier (BBB), target determination, ignorance of biodistribution after administration, and complex intercellular communication between brain cells, make brain theranostics application difficult, especially when it comes to clinical application. So, a more thorough understanding of each aspect is needed. In this review, we focus on recent studies regarding the role of exosomes in intercellular communication of brain cells, therapeutic effect of graphene quantum dots, transcriptomics/epitranscriptomics approach for target selection, and in vitro/in vivo considerations.
Collapse
Affiliation(s)
- Minseok Suh
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 03080 Republic of Korea
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 03080 Republic of Korea
| |
Collapse
|
75
|
Di Lorenzo F, Ponzo V, Motta C, Bonnì S, Picazio S, Caltagirone C, Bozzali M, Martorana A, Koch G. Impaired Spike Timing Dependent Cortico-Cortical Plasticity in Alzheimer’s Disease Patients. J Alzheimers Dis 2018; 66:983-991. [DOI: 10.3233/jad-180503] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Francesco Di Lorenzo
- Department of Behavioural and Clinical Neurology, Non-invasive Brain Stimulation Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Viviana Ponzo
- Department of Behavioural and Clinical Neurology, Non-invasive Brain Stimulation Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Caterina Motta
- Department of Behavioural and Clinical Neurology, Non-invasive Brain Stimulation Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Sonia Bonnì
- Department of Behavioural and Clinical Neurology, Non-invasive Brain Stimulation Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Silvia Picazio
- Department of Behavioural and Clinical Neurology, Non-invasive Brain Stimulation Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Carlo Caltagirone
- Department of Behavioural and Clinical Neurology, Non-invasive Brain Stimulation Unit, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of System Medicine, Tor Vergata University, Rome, Italy
| | - Marco Bozzali
- Neuroimaging Laboratory, Santa Lucia Foundation, IRCCS, Rome, Italy
| | - Alessandro Martorana
- Department of Behavioural and Clinical Neurology, Non-invasive Brain Stimulation Unit, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of System Medicine, Tor Vergata University, Rome, Italy
| | - Giacomo Koch
- Department of Behavioural and Clinical Neurology, Non-invasive Brain Stimulation Unit, Santa Lucia Foundation IRCCS, Rome, Italy
- Stroke Unit, Tor Vergata Hospital, Rome, Italy
| |
Collapse
|
76
|
Gajera CR, Fernandez R, Postupna N, Montine KS, Fox EJ, Tebaykin D, Angelo M, Bendall SC, Keene CD, Montine TJ. Mass synaptometry: High-dimensional multi parametric assay for single synapses. J Neurosci Methods 2018; 312:73-83. [PMID: 30465796 DOI: 10.1016/j.jneumeth.2018.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/09/2018] [Accepted: 11/10/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND Synaptic alterations, especially presynaptic changes, are cardinal features of neurodegenerative diseases and strongly correlate with cognitive decline. NEW METHOD We report "Mass Synaptometry" for the high-dimensional analysis of individual human synaptosomes, enriched nerve terminals from brain. This method was adapted from cytometry by time-of-flight mass spectrometry (CyTOF), which is commonly used for single-cell analysis of immune and blood cells. RESULT Here we overcome challenges for single synapse analysis by optimizing synaptosome preparations, generating a 'SynTOF panel,' recalibrating acquisition settings, and applying computational analyses. Through the analysis of 390,000 individual synaptosomes, we also provide proof-of principle validation by characterizing changes in synaptic diversity in Lewy Body Disease (LBD), Alzheimer's disease and normal brain. COMPARISON WITH EXISTING METHOD(S) Current imaging methods to study synapses in humans are capable of analyzing a limited number of synapses, and conventional flow cytometric techniques are typically restricted to fewer than 6 parameters. Our method allows for the simultaneous detection of 34 parameters from tens of thousands of individual synapses. CONCLUSION We applied Mass Synaptometry to analyze 34 parameters simultaneously on more than 390,000 synaptosomes from 13 human brain samples. This new approach revealed regional and disease-specific changes in synaptic phenotypes, including validation of this method with the expected changes in the molecular composition of striatal dopaminergic synapses in Lewy body disease and Alzheimer's disease. Mass synaptometry enables highly parallel molecular profiling of individual synaptic terminals.
Collapse
Affiliation(s)
- Chandresh R Gajera
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Rosemary Fernandez
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Nadia Postupna
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Kathleen S Montine
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Edward J Fox
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Dmitry Tebaykin
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Michael Angelo
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Sean C Bendall
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Thomas J Montine
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States.
| |
Collapse
|
77
|
Velasco-Estevez M, Mampay M, Boutin H, Chaney A, Warn P, Sharp A, Burgess E, Moeendarbary E, Dev KK, Sheridan GK. Infection Augments Expression of Mechanosensing Piezo1 Channels in Amyloid Plaque-Reactive Astrocytes. Front Aging Neurosci 2018; 10:332. [PMID: 30405400 PMCID: PMC6204357 DOI: 10.3389/fnagi.2018.00332] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/01/2018] [Indexed: 01/07/2023] Open
Abstract
A defining pathophysiological hallmark of Alzheimer's disease (AD) is the amyloid plaque; an extracellular deposit of aggregated fibrillar Aβ1-42 peptides. Amyloid plaques are hard, brittle structures scattered throughout the hippocampus and cerebral cortex and are thought to cause hyperphosphorylation of tau, neurofibrillary tangles, and progressive neurodegeneration. Reactive astrocytes and microglia envelop the exterior of amyloid plaques and infiltrate their inner core. Glia are highly mechanosensitive cells and can almost certainly sense the mismatch between the normally soft mechanical environment of the brain and very stiff amyloid plaques via mechanosensing ion channels. Piezo1, a non-selective cation channel, can translate extracellular mechanical forces to intracellular molecular signaling cascades through a process known as mechanotransduction. Here, we utilized an aging transgenic rat model of AD (TgF344-AD) to study expression of mechanosensing Piezo1 ion channels in amyloid plaque-reactive astrocytes. We found that Piezo1 is upregulated with age in the hippocampus and cortex of 18-month old wild-type rats. However, more striking increases in Piezo1 were measured in the hippocampus of TgF344-AD rats compared to age-matched wild-type controls. Interestingly, repeated urinary tract infections with Escherichia coli bacteria, a common comorbidity in elderly people with dementia, caused further elevations in Piezo1 channel expression in the hippocampus and cortex of TgF344-AD rats. Taken together, we report that aging and peripheral infection augment amyloid plaque-induced upregulation of mechanoresponsive ion channels, such as Piezo1, in astrocytes. Further research is required to investigate the role of astrocytic Piezo1 in the Alzheimer's brain, whether modulating channel opening will protect or exacerbate the disease state, and most importantly, if Piezo1 could prove to be a novel drug target for age-related dementia.
Collapse
Affiliation(s)
- María Velasco-Estevez
- Neuroimmulology & Neurotherapeutics Laboratory, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
- Drug Development, Department of Physiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Myrthe Mampay
- Neuroimmulology & Neurotherapeutics Laboratory, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Hervé Boutin
- Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, United Kingdom
| | - Aisling Chaney
- Wolfson Molecular Imaging Centre, Faculty of Biology, Medicine and Health and Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, United Kingdom
- Department of Radiology, Stanford University, Stanford, CA, United States
| | - Peter Warn
- Evotec (UK) Ltd., Manchester Science Park, Manchester, United Kingdom
| | - Andrew Sharp
- Evotec (UK) Ltd., Manchester Science Park, Manchester, United Kingdom
| | - Ellie Burgess
- Evotec (UK) Ltd., Manchester Science Park, Manchester, United Kingdom
| | - Emad Moeendarbary
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Kumlesh K. Dev
- Drug Development, Department of Physiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Graham K. Sheridan
- Neuroimmulology & Neurotherapeutics Laboratory, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| |
Collapse
|
78
|
Scholz D, Chernyshova Y, Ückert AK, Leist M. Reduced Aβ secretion by human neurons under conditions of strongly increased BACE activity. J Neurochem 2018; 147:256-274. [PMID: 29804308 DOI: 10.1111/jnc.14467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/06/2018] [Accepted: 05/23/2018] [Indexed: 12/20/2022]
Abstract
The initial step in the amyloidogenic cascade of amyloid precursor protein (APP) processing is catalyzed by beta-site APP-cleaving enzyme (BACE), and this protease has increased activities in affected areas of Alzheimer's disease brains. We hypothesized that altered APP processing, because of augmented BACE activity, would affect the actions of direct and indirect BACE inhibitors. We therefore compared post-mitotic human neurons (LUHMES) with their BACE-overexpressing counterparts (BLUHMES). Although β-cleavage of APP was strongly increased in BLUHMES, they produced less full-length and truncated amyloid beta (Aβ) than LUHMES. Moreover, low concentrations of BACE inhibitors decreased cellular BACE activity as expected, but increased Aβ1-40 levels. Several other approaches to modulate BACE activity led to a similar, apparently paradoxical, behavior. For instance, reduction in intracellular acidification by bepridil increased Aβ production in parallel with decreased BACE activity. In contrast to BLUHMES, the respective control cells (LUHMES or BLUHMES with catalytically inactive BACE) showed conventional pharmacological responses. Other non-canonical neurochemical responses (so-called 'rebound effects') are well-documented for the Aβ pathway, especially for γ-secretase: a partial block of its activity leads to an increased Aβ secretion by some cell types. We therefore compared LUHMES and BLUHMES regarding rebound effects of γ-secretase inhibitors and found an Aβ rise in LUHMES but not in BLUHMES. Thus, different cellular factors are responsible for the γ-secretase- versus BACE-related Aβ rebound. We conclude that increased BACE activity, possibly accompanied by an altered cellular localization pattern, can dramatically influence Aβ generation in human neurons and affect pharmacological responses to secretase inhibitors. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Diana Scholz
- Chair for in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Yana Chernyshova
- Chair for in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Anna-Katharina Ückert
- Chair for in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Marcel Leist
- Chair for in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| |
Collapse
|
79
|
Aliès B, Borghesani V, Noël S, Sayen S, Guillon E, Testemale D, Faller P, Hureau C. Mutations of Histidine 13 to Arginine and Arginine 5 to Glycine Are Responsible for Different Coordination Sites of Zinc(II) to Human and Murine Peptides. Chemistry 2018; 24:14233-14241. [PMID: 29978925 DOI: 10.1002/chem.201802759] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Indexed: 01/21/2023]
Abstract
Because mice and rats do not naturally develop Alzheimer's disease, genetically modified animals are required to study this pathology. This striking difference in terms of disease onset could be due to three alterations in the murine sequence (R5G, Y10F and H13R) of the amyloid-β peptide with respect to the human counterpart. Whether the metal-ion binding properties of the murine peptide are at the origin of such different amyloidogenicity of the two peptides is still an open question. Herein, the main zinc binding site to the murine amyloid-β at physiological pH has been determined through the combination of several spectroscopic and analytical methods applied to a series of six peptides with one or two of the key mutations. These results have been compared with the zinc binding site encountered in the human peptide. A coordination mechanism that demonstrates the importance of the H13R and R5G mutations in the different zinc environments present in the murine and human peptides is proposed. The nature of the minor zinc species present at physiological pH is also suggested for both peptides. Finally, the biological relevance and fallouts of the differences determined in zinc binding to human versus murine amyloid-β are also discussed.
Collapse
Affiliation(s)
- Bruno Aliès
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France.,Current address: Université de Bordeaux, ChemBioPharm INSERM U1212 CNRS UMR 5320, 33076, Bordeaux, France
| | | | - Sabrina Noël
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Stephanie Sayen
- Université Reims Champagne Ardenne, Institut de Chimie Moléculaire de Reims (ICMR), UMR 7312 CNRS-URCA, Moulin de la Housse, BP 1039, 51687, Reims Cedex 2, France
| | - Emmanuel Guillon
- Université Reims Champagne Ardenne, Institut de Chimie Moléculaire de Reims (ICMR), UMR 7312 CNRS-URCA, Moulin de la Housse, BP 1039, 51687, Reims Cedex 2, France
| | - Denis Testemale
- Univ. Grenoble Alpes, CNRS, Grenoble INP, Institut Néel, 38000, Grenoble, France.,BM30B/FAME, ESRF, The European Synchrotron, 71 avenue des Martyrs, 38000, Grenoble, France
| | - Peter Faller
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France.,Current address: Institut de Chimie, UMR 7177 CNRS-Université de Strasbourg, 4 rue Blaise Pascal, Institut Le Bel, 67008, Strasbourg, France
| | | |
Collapse
|
80
|
Blood-derived amyloid-β protein induces Alzheimer's disease pathologies. Mol Psychiatry 2018; 23:1948-1956. [PMID: 29086767 DOI: 10.1038/mp.2017.204] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 08/04/2017] [Accepted: 08/09/2017] [Indexed: 02/07/2023]
Abstract
The amyloid-β protein (Aβ) protein plays a pivotal role in the pathogenesis of Alzheimer's disease (AD). It is believed that Aβ deposited in the brain originates from the brain tissue itself. However, Aβ is generated in both brain and peripheral tissues. Whether circulating Aβ contributes to brain AD-type pathologies remains largely unknown. In this study, using a model of parabiosis between APPswe/PS1dE9 transgenic AD mice and their wild-type littermates, we observed that the human Aβ originated from transgenic AD model mice entered the circulation and accumulated in the brains of wild-type mice, and formed cerebral amyloid angiopathy and Aβ plaques after a 12-month period of parabiosis. AD-type pathologies related to the Aβ accumulation including tau hyperphosphorylation, neurodegeneration, neuroinflammation and microhemorrhage were found in the brains of the parabiotic wild-type mice. More importantly, hippocampal CA1 long-term potentiation was markedly impaired in parabiotic wild-type mice. To the best of our knowledge, our study is the first to reveal that blood-derived Aβ can enter the brain, form the Aβ-related pathologies and induce functional deficits of neurons. Our study provides novel insight into AD pathogenesis and provides evidence that supports the development of therapies for AD by targeting Aβ metabolism in both the brain and the periphery.
Collapse
|
81
|
Anti-diabetic vanadyl complexes reduced Alzheimer's disease pathology independent of amyloid plaque deposition. SCIENCE CHINA-LIFE SCIENCES 2018; 62:126-139. [PMID: 30136058 DOI: 10.1007/s11427-018-9350-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/16/2018] [Indexed: 12/20/2022]
Abstract
Association of Alzheimer's disease (AD) with cerebral glucose hypometabolism, likely due to impairments of insulin signaling, has been reported recently, with encouraging results when additional insulin is provided to AD patients. Here, we tested the potential effects of the anti-diabetic vanadium, vanadyl (IV) acetylacetonate (VAC), on AD in vitro and in vivo models. The experimental results showed that VAC at sub-micromolar concentrations improved the viability of neural cells with or without increased β-amyloid (Aβ) burden; and in APP/PS1 transgenic mice, VAC treatment (0.1 mmol kg-1 d-1) preserved cognitive function and attenuated neuron loss, but did not reduce brain Aβ plaques. Further studies revealed that VAC attenuated Aβ pathogenesis by (i) activation of the PPARγ-AMPK signal transduction pathway, leading to improved glucose and energy metabolism; (ii) up-regulation of the expression of glucose-regulated protein 75 (Grp75), thus suppressing p53-mediated neuronal apoptosis under Aβ-related stresses; and (iii) decreasing toxic soluble Aβ peptides. Overall, our work suggested that vanadyl complexes may have great potential for effective therapeutic treatment of AD.
Collapse
|
82
|
Ranjan VD, Qiu L, Tan EK, Zeng L, Zhang Y. Modelling Alzheimer's disease: Insights from in vivo to in vitro three-dimensional culture platforms. J Tissue Eng Regen Med 2018; 12:1944-1958. [PMID: 30011422 DOI: 10.1002/term.2728] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/21/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by progressive memory loss, impairment of other cognitive functions, and inability to perform activities of daily life. The key to understanding AD aetiology lies in the development of effective disease models, which should ideally recapitulate all aspects pertaining to the disease. A plethora of techniques including in vivo, in vitro, and in silico platforms have been utilized in developing disease models of AD over the years. Each of these approaches has revealed certain essential characteristics of AD; however, none have managed to fully mimic the pathological hallmarks observed in the AD human brain. In this review, we will provide details into the genesis, evolution, and significance of the principal methods currently employed in modelling AD, the advantages and limitations faced in their application, including the headways made by each approach. This review will focus primarily on two-dimensional and three-dimensional in vitro modelling of AD, which during the last few years has made significant breakthroughs in the areas of AD pathology and therapeutic screening. In addition, a glimpse into state-of-the-art neural tissue engineering techniques incorporating biomaterials and microfluidics technologies is provided, which could pave the way for the development of more accurate and comprehensive AD models in the future.
Collapse
Affiliation(s)
- Vivek Damodar Ranjan
- NTU Institute for Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, Singapore.,School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore.,Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| | - Yilei Zhang
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore
| |
Collapse
|
83
|
Kiyota T, Machhi J, Lu Y, Dyavarshetty B, Nemati M, Yokoyama I, Mosley RL, Gendelman HE. Granulocyte-macrophage colony-stimulating factor neuroprotective activities in Alzheimer's disease mice. J Neuroimmunol 2018; 319:80-92. [PMID: 29573847 PMCID: PMC5916331 DOI: 10.1016/j.jneuroim.2018.03.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/27/2018] [Accepted: 03/15/2018] [Indexed: 12/11/2022]
Abstract
We investigated the effects of granulocyte-macrophage colony stimulating factor (GM-CSF) on behavioral and pathological outcomes in Alzheimer's disease (AD) and non-transgenic mice. GM-CSF treatment in AD mice reduced brain amyloidosis, increased plasma Aβ, and rescued cognitive impairment with increased hippocampal expression of calbindin and synaptophysin and increased levels of doublecortin-positive cells in the dentate gyrus. These data extend GM-CSF pleiotropic neuroprotection mechanisms in AD and include regulatory T cell-mediated immunomodulation of microglial function, Aβ clearance, maintenance of synaptic integrity, and induction of neurogenesis. Together these data support further development of GM-CSF as a neuroprotective agent for AD.
Collapse
Affiliation(s)
- Tomomi Kiyota
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bhagyalaxmi Dyavarshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maryam Nemati
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Izumi Yokoyama
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - R L Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
84
|
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative cortical dementia. It starts with memory loss, spatial disorientation in people above the age of 65 yr with a preference to females. Its incidence is expected to increase threefold by 2050. It affects almost one out of ten persons above the age of 65 years. Majority of patients are sporadic, but a very small percentage is autosomal dominant. The pathomechanisms postulated include amyloid cascade hypothesis according to which mutation in amyloid precursor protein causes Aβ aggregation. The next hypothesis is signal transducer and activation of transcription 3 (STAT3) causing aberration in intracellular signalling pathways. Senile plaques and neurofibrillary tangles are other important pathological changes reported. It is observed that dementia research has not yielded the expected result world over, and therefore, the pitfalls with reference to known facts about diagnosis, clinical features, pathogenic mechanisms, assessment of progression, biomarkers, treatment and prevention, as well as brief information on our experiments with relatively inexpensive methods of differentiating the most common types of dementia AD and frontotemporal dementia are discussed.
Collapse
|
85
|
Development of Microplatforms to Mimic the In Vivo Architecture of CNS and PNS Physiology and Their Diseases. Genes (Basel) 2018; 9:genes9060285. [PMID: 29882823 PMCID: PMC6027402 DOI: 10.3390/genes9060285] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 12/16/2022] Open
Abstract
Understanding the mechanisms that govern nervous tissues function remains a challenge. In vitro two-dimensional (2D) cell culture systems provide a simplistic platform to evaluate systematic investigations but often result in unreliable responses that cannot be translated to pathophysiological settings. Recently, microplatforms have emerged to provide a better approximation of the in vivo scenario with better control over the microenvironment, stimuli and structure. Advances in biomaterials enable the construction of three-dimensional (3D) scaffolds, which combined with microfabrication, allow enhanced biomimicry through precise control of the architecture, cell positioning, fluid flows and electrochemical stimuli. This manuscript reviews, compares and contrasts advances in nervous tissues-on-a-chip models and their applications in neural physiology and disease. Microplatforms used for neuro-glia interactions, neuromuscular junctions (NMJs), blood-brain barrier (BBB) and studies on brain cancer, metastasis and neurodegenerative diseases are addressed. Finally, we highlight challenges that can be addressed with interdisciplinary efforts to achieve a higher degree of biomimicry. Nervous tissue microplatforms provide a powerful tool that is destined to provide a better understanding of neural health and disease.
Collapse
|
86
|
Wang P, Zheng X, Guo Q, Yang P, Pang X, Qian K, Lu W, Zhang Q, Jiang X. Systemic delivery of BACE1 siRNA through neuron-targeted nanocomplexes for treatment of Alzheimer's disease. J Control Release 2018; 279:220-233. [DOI: 10.1016/j.jconrel.2018.04.034] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/22/2018] [Accepted: 04/17/2018] [Indexed: 10/17/2022]
|
87
|
Pistollato F, Iglesias RC, Ruiz R, Aparicio S, Crespo J, Lopez LD, Manna PP, Giampieri F, Battino M. Nutritional patterns associated with the maintenance of neurocognitive functions and the risk of dementia and Alzheimer’s disease: A focus on human studies. Pharmacol Res 2018; 131:32-43. [DOI: 10.1016/j.phrs.2018.03.012] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 11/25/2022]
|
88
|
Zhou FQ, Jiang J, Griffith CM, Patrylo PR, Cai H, Chu Y, Yan XX. Lack of human-like extracellular sortilin neuropathology in transgenic Alzheimer's disease model mice and macaques. Alzheimers Res Ther 2018; 10:40. [PMID: 29690919 PMCID: PMC5978992 DOI: 10.1186/s13195-018-0370-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/19/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a devastating neurodegenerative disorder bearing multiple pathological hallmarks suggestive of complex cellular/molecular interplay during pathogenesis. Transgenic mice and nonhuman primates are used as disease models for mechanistic and translational research into AD; the extent to which these animal models recapitulate AD-type neuropathology is an issue of importance. Putative C-terminal fragments from sortilin, a member of the vacuolar protein sorting 10 protein (Vps10p) family, have recently been shown to deposit in the neuritic β-amyloid (Aβ) plaques in the human brain. METHODS We set out to explore if extracellular sortilin neuropathology exists in AD-related transgenic mice and nonhuman primates. Brains from different transgenic strains and ages developed overt cerebral Aβ deposition, including the β-amyloid precursor protein and presenilin 1 double-transgenic (APP/PS1) mice at ~ 14 months of age, the five familial Alzheimer's disease mutations transgenic (5×FAD) mice at ~ 8 months, the triple-transgenic Alzheimer's disease (3×Tg-AD) mice at ~ 22 months, and aged monkeys (Macaca mulatta and Macaca fascicularis) were examined. Brain samples from young transgenic mice, middle-aged/aged monkeys, and AD humans were used as negative and positive pathological controls. RESULTS The C-terminal sortilin antibody, which labeled senile plaques in the AD human cerebral sections, did not display extracellular immunolabeling in the transgenic mouse or aged monkey brain sections with Aβ deposition. In Western blot analysis, sortilin fragments ~ 15 kDa were not detectable in transgenic mouse cortical lysates, but they occurred in control AD lysates. CONCLUSIONS In reference to their human brain counterparts, neuritic plaques seen in transgenic AD model mouse brains represent an incomplete form of this AD pathological hallmark. The species difference in neuritic plaque constituents also indicates more complex secondary proteopathies in the human brain relative to rodents and nonhuman primates during aging and in AD.
Collapse
Affiliation(s)
- Feng-Qin Zhou
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, 410013 Hunan China
| | - Juan Jiang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, 410013 Hunan China
| | - Chelsea M. Griffith
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901 USA
| | - Peter R. Patrylo
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901 USA
| | - Huaibin Cai
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892 USA
| | - Yaping Chu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612 USA
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, 410013 Hunan China
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, Hunan China
| |
Collapse
|
89
|
Transgenic autoinhibition of p21-activated kinase exacerbates synaptic impairments and fronto-dependent behavioral deficits in an animal model of Alzheimer's disease. Aging (Albany NY) 2018; 9:1386-1403. [PMID: 28522792 PMCID: PMC5472739 DOI: 10.18632/aging.101239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/11/2017] [Indexed: 11/28/2022]
Abstract
Defects in p21-activated kinase (PAK) lead to dendritic spine abnormalities and are sufficient to cause cognition impairment. The decrease in PAK in the brain of Alzheimer's disease (AD) patients is suspected to underlie synaptic and dendritic disturbances associated with its clinical expression, particularly with symptoms related to frontal cortex dysfunction. To investigate the role of PAK combined with Aβ and tau pathologies (3xTg-AD mice) in the frontal cortex, we generated a transgenic model of AD with a deficit in PAK activity (3xTg-AD-dnPAK mice). PAK inactivation had no effect on Aβ40 and Aβ42 levels, but increased the phosphorylation ratio of tau in detergent-insoluble protein fractions in the frontal cortex of 18-month-old heterozygous 3xTg-AD mice. Morphometric analyses of layer II/III pyramidal neurons in the frontal cortex showed that 3xTg-AD-dnPAK neurons exhibited significant dendritic attrition, lower spine density and longer spines compared to NonTg and 3xTg-AD mice. Finally, behavioral assessments revealed that 3xTg-AD-dnPAK mice exhibited pronounced anxious traits and disturbances in social behaviors, reminiscent of fronto-dependent symptoms observed in AD. Our results substantiate a critical role for PAK in the genesis of neuronal abnormalities in the frontal cortex underlying the emergence of psychiatric-like symptoms in AD.
Collapse
|
90
|
Le Duigou C, Savary E, Morin-Brureau M, Gomez-Dominguez D, Sobczyk A, Chali F, Milior G, Kraus L, Meier JC, Kullmann DM, Mathon B, de la Prida LM, Dorfmuller G, Pallud J, Eugène E, Clemenceau S, Miles R. Imaging pathological activities of human brain tissue in organotypic culture. J Neurosci Methods 2018; 298:33-44. [PMID: 29427611 DOI: 10.1016/j.jneumeth.2018.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/27/2018] [Accepted: 02/02/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Insights into human brain diseases may emerge from tissue obtained after operations on patients. However techniques requiring transduction of transgenes carried by viral vectors cannot be applied to acute human tissue. NEW METHOD We show that organotypic culture techniques can be used to maintain tissue from patients with three different neurological syndromes for several weeks in vitro. Optimized viral vector techniques and promoters for transgene expression are described. RESULTS Region-specific differences in neuronal form, firing pattern and organization as well as pathological activities were maintained over 40-50 days in culture. Both adeno-associated virus and lentivirus based vectors were persistently expressed from ∼10 days after application, providing 30-40 days to exploit genetically expressed constructs. Different promoters, including hSyn, e/hSyn, CMV and CaMKII, provided cell-type specific transgene expression. The Ca probe GCaMP let us explore epileptogenic synchrony and a FRET-based probe was used to follow activity of the kinase mTORC1. COMPARISON WITH EXISTING METHODS The use of a defined culture medium, with low concentrations of amino acids and no growth factors, permitted organotypic culture of tissue from humans aged 3-62 years. Epileptic activity was maintained and excitability changed relatively little until ∼6 weeks in culture. CONCLUSIONS Characteristic morphology and region-specific neuronal activities are maintained in organotypic culture of tissue from patients diagnosed with mesial temporal lobe epilepsy, cortical dysplasia and cortical glioblastoma. Viral vector techniques permit expression of probes for long-term measurements of multi-cellular activity and intra-cellular signaling.
Collapse
Affiliation(s)
- Caroline Le Duigou
- Cortex & Epilepsie, Inserm U1127, CNRS UMR7225, UPMC Univ Paris 6, Institut du Cerveau et de la Moelle épinière, Paris, 75013, France, France.
| | - Etienne Savary
- Cortex & Epilepsie, Inserm U1127, CNRS UMR7225, UPMC Univ Paris 6, Institut du Cerveau et de la Moelle épinière, Paris, 75013, France, France.
| | - Mélanie Morin-Brureau
- Cortex & Epilepsie, Inserm U1127, CNRS UMR7225, UPMC Univ Paris 6, Institut du Cerveau et de la Moelle épinière, Paris, 75013, France, France
| | - Daniel Gomez-Dominguez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, E-28002, Spain
| | - André Sobczyk
- Cortex & Epilepsie, Inserm U1127, CNRS UMR7225, UPMC Univ Paris 6, Institut du Cerveau et de la Moelle épinière, Paris, 75013, France, France
| | - Farah Chali
- Cortex & Epilepsie, Inserm U1127, CNRS UMR7225, UPMC Univ Paris 6, Institut du Cerveau et de la Moelle épinière, Paris, 75013, France, France
| | - Giampaolo Milior
- Cortex & Epilepsie, Inserm U1127, CNRS UMR7225, UPMC Univ Paris 6, Institut du Cerveau et de la Moelle épinière, Paris, 75013, France, France
| | - Larissa Kraus
- Cell Physiology, Technische Universität Braunschweig, Braunschweig, Germany; Charite Universitätsmedizin, Clinical and Experimental Epileptology, Berlin, Germany; Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Jochen C Meier
- Cell Physiology, Technische Universität Braunschweig, Braunschweig, Germany
| | | | - Bertrand Mathon
- Neurochirurgie, AP-HP, GH Pitie-Salpêtrière-Charles Foix, Paris, 75013, France
| | | | - Georg Dorfmuller
- Neurochirurgie, Fondation Ophtalmologique Rothschild, 75019, Paris, France
| | - Johan Pallud
- Neurochirurgie, Hôpital Sainte-Anne, Paris Descartes University, IMA-BRAIN, Inserm, U894 Centre de Psychiatrie et Neurosciences, Paris, 75014, France
| | - Emmanuel Eugène
- Inserm U839, UPMC Univ Paris 6, Institut du Fer-à-Moulin, Paris, 75005, France
| | - Stéphane Clemenceau
- Neurochirurgie, AP-HP, GH Pitie-Salpêtrière-Charles Foix, Paris, 75013, France
| | - Richard Miles
- Cortex & Epilepsie, Inserm U1127, CNRS UMR7225, UPMC Univ Paris 6, Institut du Cerveau et de la Moelle épinière, Paris, 75013, France, France.
| |
Collapse
|
91
|
McGraw CM, Ward CS, Samaco RC. Genetic rodent models of brain disorders: Perspectives on experimental approaches and therapeutic strategies. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2018; 175:368-379. [PMID: 28910526 PMCID: PMC5659732 DOI: 10.1002/ajmg.c.31570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 06/28/2017] [Indexed: 12/14/2022]
Abstract
Neurobehavioral disorders comprised of neurodegenerative, neurodevelopmental, and psychiatric disorders together represent leading causes of morbidity and mortality. Despite significant academic research and industry efforts to elucidate the disease mechanisms operative in these disorders and to develop mechanism‐based therapies, our understanding remains incomplete and our access to tractable therapeutic interventions severely limited. The magnitude of these short‐comings can be measured by the growing list of disappointing clinical trials based on initially promising compounds identified in genetic animal models. This review and commentary will explore why this may be so, focusing on the central role that genetic models of neurobehavioral disorders have come to occupy in current efforts to identify disease mechanisms and therapies. In particular, we will highlight the unique pitfalls and challenges that have hampered success in these models as compared to genetic models of non‐neurological diseases as well as to symptom‐based models of the early 20th century that led to the discovery of all major classes of psychoactive pharmaceutical compounds still used today. Using examples from specific genetic rodent models of human neurobehavioral disorders, we will highlight issues of reproducibility, construct validity, and translational relevance in the hopes that these examples will be instructive toward greater success in future endeavors. Lastly, we will champion a two‐pronged approach toward identifying novel therapies for neurobehavioral disorders that makes greater use of the historically more successful symptom‐based approaches in addition to more mechanism‐based approaches.
Collapse
Affiliation(s)
- Christopher M McGraw
- Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Christopher S Ward
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Rodney C Samaco
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
92
|
Ahlemeyer B, Halupczok S, Rodenberg-Frank E, Valerius KP, Baumgart-Vogt E. Endogenous Murine Amyloid-β Peptide Assembles into Aggregates in the Aged C57BL/6J Mouse Suggesting These Animals as a Model to Study Pathogenesis of Amyloid-β Plaque Formation. J Alzheimers Dis 2018; 61:1425-1450. [DOI: 10.3233/jad-170923] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Barbara Ahlemeyer
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Giessen, Germany
| | - Sascha Halupczok
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Giessen, Germany
| | - Elke Rodenberg-Frank
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Giessen, Germany
| | - Klaus-Peter Valerius
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology, Division of Medical Cell Biology, Justus Liebig University, Giessen, Germany
| |
Collapse
|
93
|
Souchet B, Audrain M, Billoir B, Lecanu L, Tada S, Braudeau J. Is it time to rethink the Alzheimer's disease drug development strategy by targeting its silent phase? Neural Regen Res 2018; 13:224-225. [PMID: 29557364 PMCID: PMC5879886 DOI: 10.4103/1673-5374.226389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
| | | | | | | | - Satoru Tada
- AgenT, 4 rue Pierre-Fontaine, 91058 EVRY Cedex, France
| | | |
Collapse
|
94
|
Sharma HS, Muresanu DF, Lafuente JV, Patnaik R, Tian ZR, Ozkizilcik A, Castellani RJ, Mössler H, Sharma A. Co-Administration of TiO2 Nanowired Mesenchymal Stem Cells with Cerebrolysin Potentiates Neprilysin Level and Reduces Brain Pathology in Alzheimer's Disease. Mol Neurobiol 2018; 55:300-311. [PMID: 28844104 DOI: 10.1007/s12035-017-0742-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Neprilysin (NPL), the rate-limiting enzyme for amyloid beta peptide (AβP), appears to play a crucial role in the pathogenesis of Alzheimer's disease (AD). Since mesenchymal stem cells (MSCs) and/or cerebrolysin (CBL, a combination of neurotrophic factors and active peptide fragments) have neuroprotective effects in various CNS disorders, we examined nanowired delivery of MSCs and CBL on NPL content and brain pathology in AD using a rat model. AD-like symptoms were produced by intraventricular (i.c.v.) administration of AβP (1-40) in the left lateral ventricle (250 ng/10 μl, once daily) for 4 weeks. After 30 days, the rats were examined for NPL and AβP concentrations in the brain and related pathology. Co-administration of TiO2-nanowired MSCs (106 cells) with 2.5 ml/kg CBL (i.v.) once daily for 1 week after 2 weeks of AβP infusion significantly increased the NPL in the hippocampus (400 pg/g) from the untreated control group (120 pg/g; control 420 ± 8 pg/g brain) along with a significant decrease in the AβP deposition (45 pg/g from untreated control 75 pg/g; saline control 40 ± 4 pg/g). Interestingly, these changes were much less evident when the MSCs or CBL treatment was given alone. Neuronal damages, gliosis, and myelin vesiculation were also markedly reduced by the combined treatment of TiO2, MSCs, and CBL in AD. These observations are the first to show that co-administration of TiO2-nanowired CBL and MSCs has superior neuroprotective effects in AD probably due to increasing the brain NPL level effectively, not reported earlier.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, SE-75185, Uppsala, Sweden.
- International Experimental Central Nervous System Injury & Repair (IECNSIR), University Hospital, Uppsala University, Frödingsgatan 12, Bldg. 28, SE-75421, Uppsala, Sweden.
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania.
- Department of Neurosciences, University of Basque Country, Bilbao, Spain.
| | - Dafin Fior Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, 37 Mircea Eliade Street, 400364, Cluj-Napoca, Romania
| | - José Vicente Lafuente
- Department of Neurosciences, University of Basque Country, Bilbao, Spain
- Nanoneurosurgery Group, BioCruces Health Research Institute, 48903, Barakaldo, Bizkaia, Spain
- Faculty of Health Science, Universidad Autónoma de Chile, Santiago de Chile, Chile
| | - Ranjana Patnaik
- School of Biomedical Engineering, Department of Biomaterials, Indian Institute of technology, Banaras Hindu University, Varanasi, India
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - Asya Ozkizilcik
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | | | - Herbert Mössler
- "RoNeuro" Institute for Neurological Research and Diagnostic, 37 Mircea Eliade Street, 400364, Cluj-Napoca, Romania
| | - Aruna Sharma
- Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, SE-75185, Uppsala, Sweden
- International Experimental Central Nervous System Injury & Repair (IECNSIR), University Hospital, Uppsala University, Frödingsgatan 12, Bldg. 28, SE-75421, Uppsala, Sweden
- "RoNeuro" Institute for Neurological Research and Diagnostic, 37 Mircea Eliade Street, 400364, Cluj-Napoca, Romania
- Department of Neurosciences, University of Basque Country, Bilbao, Spain
| |
Collapse
|
95
|
Bai H, Zhang QF, Duan JJ, Yu DJ, Liu LJ. Downregulation of signal transduction and STAT3 expression exacerbates oxidative stress mediated by NLRP3 inflammasome. Neural Regen Res 2018; 13:2147-2155. [PMID: 30323145 PMCID: PMC6199955 DOI: 10.4103/1673-5374.241470] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Activated nucleotide binding to the oligonucleotide receptor protein 3 (NLRP3) inflammasome is possibly involved in the pathogenesis of Alzheimer's disease through oxidative stress and neurogenic inflammation. Low expression of the signal transducer and activator of transcription 3 (STAT3) gene may promote the occurrence of neurodegenerative diseases to some extent. To clarify the roles of the NLRP3 inflammasome and STAT3 expression in oxidative stress, (1) SHSY5Y cells were incubated with 1 mM H2O2 to induce oxidative stress injury, and the expression of human-cell-specific signal transduction, STAT3-shRNA silencing signal transduction and STAT3 were detected. Cells were pretreated with Ca2+ chelator BAPATA-AM (0.1 mM) for 30 minutes as a control. (2) Western blot assay was used to analyze the expression of caspase-1, NLRP3, signal transduction and STAT3. Enzyme-linked immunosorbent assay was used to analyze interleukin-1β levels. Flow cytometry was carried out to calculate the number of apoptotic cells. We found that H2O2 treatment activated NLRP3 inflammasomes and decreased phosphorylation of signal transduction and STAT3 serine 727. BAPTA-AM pretreatment abolished the H2O2-induced activation of NLRP3 inflammasomes, caspase-1 expression, interleukin-1β expression and apoptosis in SHSY5Y cells, and had no effect in cells with downregulated STAT3 expression by RNAi. The findings suggest that downregulation of signal transduction and STAT3 expression may enhance the oxidative stress mediated by NLRP3, which may not depend on the Ca2+ signaling pathway.
Collapse
Affiliation(s)
- Hua Bai
- Medical Laboratory Center; Department of Neurology, Third Affiliated Hospital, Guizhou Medical University, Duyun, Guizhou Province, China
| | - Qi-Fang Zhang
- Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, and Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Juan-Juan Duan
- Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, and Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - De-Jun Yu
- Medical Laboratory Center, Third Affiliated Hospital, Guizhou Medical University, Duyun, Guizhou Province, China
| | - Li-Jie Liu
- Department of Neurology, Third Affiliated Hospital, Guizhou Medical University, Duyun, Guizhou Province, China
| |
Collapse
|
96
|
Fessel J. Amyloid is essential but insufficient for Alzheimer causation: addition of subcellular cofactors is required for dementia. Int J Geriatr Psychiatry 2018; 33:e14-e21. [PMID: 28509380 DOI: 10.1002/gps.4730] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 04/03/2017] [Indexed: 01/14/2023]
Abstract
OBJECTIVE The aim of this study is to examine the hypotheses stating the importance of amyloid or of its oligomers in the pathogenesis of Alzheimer's disease (AD). METHODS Published studies were examined. RESULTS The importance of amyloid in the pathogenesis of AD is well established, yet accepting it as the main cause for AD is problematic, because amyloid-centric treatments have provided no clinical benefit and about one-third of cognitively normal, older persons have cerebral amyloid plaques. Also problematic is the alternative hypothesis that, instead of amyloid plaques, it is oligomers of amyloid precursor protein that cause AD.Evidence is presented suggesting amyloid/oligomers as necessary but insufficient causes of the dementia and that, for dementia to develop, requires the addition of cofactors known to be associated with AD. Those cofactors include several subcellular processes: mitochondrial impairments; the Wnt signaling system; the unfolded protein response; the ubiquitin proteasome system; the Notch signaling system; and tau, calcium, and oxidative damage. CONCLUSIONS A modified amyloid/oligomer hypothesis for the pathogenesis of AD is that activation of one or more of the aforementioned cofactors creates a burden of functional impairments that, in conjunction with amyloid/oligomers, now crosses a threshold of dysfunction that results in clinical dementia. Of considerable importance, several treatments that might reverse the activation of some of the subcellular processes are available, for example, lithium, pioglitazone, erythropoietin, and prazosin; they should be given in combination in a clinical trial to test their safety and efficacy. © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Clinical Trials Unit, Kaiser Permanente, San Francisco, CA, USA
| |
Collapse
|
97
|
Xiao-Li Z, Meng-Bei X, Ting-Yu J, Pei-Qing R, Guo-Qing Z, Yan L. Preclinical Evidence and Possible Mechanisms of Extracts or Compounds from Cistanches for Alzheimer’s Disease. Aging Dis 2018. [DOI: 10.14336/ad.2018.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
98
|
Levy Nogueira M, Hamraz M, Abolhassani M, Bigan E, Lafitte O, Steyaert J, Dubois B, Schwartz L. Mechanical stress increases brain amyloid β, tau, and α‐synuclein concentrations in wild‐type mice. Alzheimers Dement 2017; 14:444-453. [DOI: 10.1016/j.jalz.2017.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 06/19/2017] [Accepted: 11/13/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Marcel Levy Nogueira
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie Hôpital de la Pitié‐Salpêtrière, AP‐HP Paris France
- Institut de Recherche Translationnelle en Neurosciences (IHU‐A‐ICM) Institut du Cerveau et de la Moelle Epinière (ICM) Paris France
| | | | | | - Erwan Bigan
- Laboratoire d'informatique (LIX), UMR 7161, École Polytechnique Université Paris‐Saclay Palaiseau France
| | - Olivier Lafitte
- LAGA, UMR 7539 Université Paris 13, Sorbonne Paris Cité Villetaneuse France
| | - Jean‐Marc Steyaert
- Laboratoire d'informatique (LIX), UMR 7161, École Polytechnique Université Paris‐Saclay Palaiseau France
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie Hôpital de la Pitié‐Salpêtrière, AP‐HP Paris France
- Institut de Recherche Translationnelle en Neurosciences (IHU‐A‐ICM) Institut du Cerveau et de la Moelle Epinière (ICM) Paris France
- INSERM, CNRS, UMR‐S975 Institut du Cerveau et de la Moelle Epinière (ICM) Paris France
| | | |
Collapse
|
99
|
LMD proteomics provides evidence for hippocampus field-specific motor protein abundance changes with relevance to Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:703-714. [DOI: 10.1016/j.bbapap.2017.03.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/17/2017] [Accepted: 03/30/2017] [Indexed: 12/13/2022]
|
100
|
Khan A, Corbett A, Ballard C. Emerging amyloid and tau targeting treatments for Alzheimer’s disease. Expert Rev Neurother 2017; 17:697-711. [DOI: 10.1080/14737175.2017.1326819] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Ayesha Khan
- Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Anne Corbett
- King’s College London, Wolfson Centre for Age-Related Diseases, London, UK
| | - Clive Ballard
- King’s College London, Wolfson Centre for Age-Related Diseases, London, UK
| |
Collapse
|