151
|
Miya Shaik M, Tamargo IA, Abubakar MB, Kamal MA, Greig NH, Gan SH. The Role of microRNAs in Alzheimer's Disease and Their Therapeutic Potentials. Genes (Basel) 2018; 9:genes9040174. [PMID: 29561798 PMCID: PMC5924516 DOI: 10.3390/genes9040174] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/05/2018] [Accepted: 03/05/2018] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are short, endogenous, non-coding RNAs that post-transcriptionally regulate gene expression by base pairing with mRNA targets. Altered miRNA expression profiles have been observed in several diseases, including neurodegeneration. Multiple studies have reported altered expressions of miRNAs in the brains of individuals with Alzheimer’s disease (AD) as compared to those of healthy elderly adults. Some of the miRNAs found to be dysregulated in AD have been reported to correlate with neuropathological changes, including plaque and tangle accumulation, as well as altered expressions of species that are known to be involved in AD pathology. To examine the potentially pathogenic functions of several dysregulated miRNAs in AD, we review the current literature with a focus on the activities of ten miRNAs in biological pathways involved in AD pathogenesis. Comprehensive understandings of the expression profiles and activities of these miRNAs will illuminate their roles as potential therapeutic targets in AD brain and may lead to the discovery of breakthrough treatment strategies for AD.
Collapse
Affiliation(s)
- Munvar Miya Shaik
- School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia.
| | - Ian A Tamargo
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Murtala B Abubakar
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, PMB 2254 Sokoto, Nigeria.
| | - Mohammad A Kamal
- Metabolomics and Enzymology Unit, Fundamental and Applied Biology Group, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Nigel H Greig
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia.
| |
Collapse
|
152
|
Machaalani R, Chen H. Brain derived neurotrophic factor (BDNF), its tyrosine kinase receptor B (TrkB) and nicotine. Neurotoxicology 2018; 65:186-195. [DOI: 10.1016/j.neuro.2018.02.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/21/2018] [Accepted: 02/25/2018] [Indexed: 02/07/2023]
|
153
|
Martin AA, Richmond JE. The sarco(endo)plasmic reticulum calcium ATPase SCA-1 regulates the Caenorhabditis elegans nicotinic acetylcholine receptor ACR-16. Cell Calcium 2018; 72:104-115. [PMID: 29748129 DOI: 10.1016/j.ceca.2018.02.005] [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: 01/24/2018] [Revised: 02/22/2018] [Accepted: 02/22/2018] [Indexed: 12/26/2022]
Abstract
Nicotinic acetylcholine receptors (nAChR) are present in many excitable tissues and are found both pre and post-synaptically. Through their non-specific cationic permeability, these nAChRs have excitatory roles in neurotransmission, neuromodulation, synaptic plasticity, and neuroprotection. Thus, nAChR mislocalization or functional deficits are associated with many neurological disease states. Therefore identifying the mechanisms that regulate nAChR expression and function will inform our understanding of normal as well as pathological physiological conditions and offer avenues for potential therapeutic advances. Taking advantage of the genetic tractability of the soil nematode Caenorhabditis elegans, a forward genetic screen was performed to isolate regulators of the vertebrate α7 nAChR homologue ACR-16. From this screen a novel regulator of the ACR-16 receptor was identified, the sarco(endo)plasmic reticulum calcium ATPase sca-1. The sca-1 mutant affects ACR-16 receptor level at the NMJ, receptor functionality, and synaptic transmission. Responses to pressure-ejected nicotine in sca-1 mutants are indistinguishable from wild type, which implies the ACR-16 receptors are mislocalized at the NMJ. Changes in cytosolic baseline calcium levels in sca-1 and other mutants indicates a calcium-driven regulation mechanism of the α7-like NAChR ACR-16.
Collapse
Affiliation(s)
- Ashley A Martin
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, United States.
| | - Janet E Richmond
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, United States
| |
Collapse
|
154
|
Rajmohan R, Reddy PH. Amyloid-Beta and Phosphorylated Tau Accumulations Cause Abnormalities at Synapses of Alzheimer's disease Neurons. J Alzheimers Dis 2018; 57:975-999. [PMID: 27567878 DOI: 10.3233/jad-160612] [Citation(s) in RCA: 378] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyloid-beta (Aβ) and hyperphosphorylated tau are hallmark lesions of Alzheimer's disease (AD). However, the loss of synapses and dysfunctions of neurotransmission are more directly tied to disease severity. The role of these lesions in the pathoetiological progression of the disease remains contested. Biochemical, cellular, molecular, and pathological studies provided several lines of evidence and improved our understanding of how Aβ and hyperphosphorylated tau accumulation may directly harm synapses and alter neurotransmission. In vitro evidence suggests that Aβ and hyperphosphorylated tau have both direct and indirect cytotoxic effects that affect neurotransmission, axonal transport, signaling cascades, organelle function, and immune response in ways that lead to synaptic loss and dysfunctions in neurotransmitter release. Observations in preclinical models and autopsy studies support these findings, suggesting that while the pathoetiology of positive lesions remains elusive, their removal may reduce disease severity and progression. The purpose of this article is to highlight the need for further investigation of the role of tau in disease progression and its interactions with Aβ and neurotransmitters alike.
Collapse
Affiliation(s)
- Ravi Rajmohan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Speech, Language and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
155
|
Niu G, Guo J, Tian Y, Zhao K, Li J, Xiao Q. α‑lipoic acid can greatly alleviate the toxic effect of AGES on SH‑SY5Y cells. Int J Mol Med 2018; 41:2855-2864. [PMID: 29436603 DOI: 10.3892/ijmm.2018.3477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 01/22/2018] [Indexed: 11/05/2022] Open
Abstract
The aim of the study was to explore the influence of α‑lipoic acid (α‑LA) on the cytotoxicity of advanced glycation end‑products (AGEs) against SH‑SY5Y cells. AGE‑bovine serum albumin (BSA) was incubated in vitro using SH‑SY5Y cells as a target model, and the control group was set. Cells were exposed to AGE‑BSA, and α‑LA was selectively added to the cells. Cell growth and death was determined by the MTT assay, which measures cellular metabolic rate, lactate dehydrogenase (LDH) leakage rate and cellular axonal length. Immunocytochemistry was employed to detect the expression of β‑amyloid (Aβ) protein in cells, and mRNA expression of amyloid precursor protein (APP) and the receptor for AGE (RAGE) were assayed by PT‑PCR. The metabolism of MTT was clearly increased, the rate of LDH leakage was significantly decreased, and axonal length was significantly increased in cells treated with α‑LA (0.1 g/l) as compared to untreated cells. Furthermore, the expression levels of Aβ protein were also decreased. In addition, α‑LA (0.1 g/l) markedly inhibited the expression of RAGE mRNA, and did not influence APP mRNA expression as compared the control group. α‑LA (0.1 g/l) was effective at dampening the cytotoxicity of AGE‑BSA, a preliminary observation that confirms the ability of α‑LA to significantly alleviate the cytotoxicity of AGEs against SH‑SY5Y cells.
Collapse
Affiliation(s)
- Guifen Niu
- Department of Endocrinology, Liaocheng City People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Jianfei Guo
- Department of Endocrinology, Liaocheng City People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Yaqiang Tian
- Department of Endocrinology, Liaocheng Brain Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Kexiang Zhao
- Department of the Elderly, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jian Li
- Department of Endocrinology, Liaocheng City People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Qian Xiao
- Department of the Elderly, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
156
|
Nakaizumi K, Ouchi Y, Terada T, Yoshikawa E, Kakimoto A, Isobe T, Bunai T, Yokokura M, Suzuki K, Magata Y. In vivo Depiction of α7 Nicotinic Receptor Loss for Cognitive Decline in Alzheimer’s Disease. J Alzheimers Dis 2018; 61:1355-1365. [DOI: 10.3233/jad-170591] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kyoko Nakaizumi
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasuomi Ouchi
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tatsuhiro Terada
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Etsuji Yoshikawa
- Central Research Laboratory, Hamamatsu Photonics K.K., Hamamatsu, Japan
| | - Akihiro Kakimoto
- Central Research Laboratory, Hamamatsu Photonics K.K., Hamamatsu, Japan
| | - Takashi Isobe
- Central Research Laboratory, Hamamatsu Photonics K.K., Hamamatsu, Japan
| | - Tomoyasu Bunai
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masamichi Yokokura
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Katsuaki Suzuki
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Kohseikai Ogasa Hospital, Kakegawa, Japan
| | - Yasuhiro Magata
- Department of Molecular Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
157
|
Forest KH, Alfulaij N, Arora K, Taketa R, Sherrin T, Todorovic C, Lawrence JLM, Yoshikawa GT, Ng HL, Hruby VJ, Nichols RA. Protection against β-amyloid neurotoxicity by a non-toxic endogenous N-terminal β-amyloid fragment and its active hexapeptide core sequence. J Neurochem 2017; 144:201-217. [PMID: 29164616 DOI: 10.1111/jnc.14257] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/01/2017] [Accepted: 11/07/2017] [Indexed: 11/28/2022]
Abstract
High levels (μM) of beta amyloid (Aβ) oligomers are known to trigger neurotoxic effects, leading to synaptic impairment, behavioral deficits, and apoptotic cell death. The hydrophobic C-terminal domain of Aβ, together with sequences critical for oligomer formation, is essential for this neurotoxicity. However, Aβ at low levels (pM-nM) has been shown to function as a positive neuromodulator and this activity resides in the hydrophilic N-terminal domain of Aβ. An N-terminal Aβ fragment (1-15/16), found in cerebrospinal fluid, was also shown to be a highly active neuromodulator and to reverse Aβ-induced impairments of long-term potentiation. Here, we show the impact of this N-terminal Aβ fragment and a shorter hexapeptide core sequence in the Aβ fragment (Aβcore: 10-15) to protect or reverse Aβ-induced neuronal toxicity, fear memory deficits and apoptotic death. The neuroprotective effects of the N-terminal Aβ fragment and Aβcore on Aβ-induced changes in mitochondrial function, oxidative stress, and apoptotic neuronal death were demonstrated via mitochondrial membrane potential, live reactive oxygen species, DNA fragmentation and cell survival assays using a model neuroblastoma cell line (differentiated NG108-15) and mouse hippocampal neuron cultures. The protective action of the N-terminal Aβ fragment and Aβcore against spatial memory processing deficits in amyloid precursor protein/PSEN1 (5XFAD) mice was demonstrated in contextual fear conditioning. Stabilized derivatives of the N-terminal Aβcore were also shown to be fully protective against Aβ-triggered oxidative stress. Together, these findings indicate an endogenous neuroprotective role for the N-terminal Aβ fragment, while active stabilized N-terminal Aβcore derivatives offer the potential for therapeutic application.
Collapse
Affiliation(s)
- Kelly H Forest
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Naghum Alfulaij
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Komal Arora
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Ruth Taketa
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Tessi Sherrin
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Cedomir Todorovic
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - James L M Lawrence
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Gene T Yoshikawa
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Ho-Leung Ng
- Department of Chemistry, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Victor J Hruby
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, USA
| | - Robert A Nichols
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
158
|
Dannenberg H, Young K, Hasselmo M. Modulation of Hippocampal Circuits by Muscarinic and Nicotinic Receptors. Front Neural Circuits 2017; 11:102. [PMID: 29321728 PMCID: PMC5733553 DOI: 10.3389/fncir.2017.00102] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 11/27/2017] [Indexed: 01/02/2023] Open
Abstract
This article provides a review of the effects of activation of muscarinic and nicotinic receptors on the physiological properties of circuits in the hippocampal formation. Previous articles have described detailed computational hypotheses about the role of cholinergic neuromodulation in enhancing the dynamics for encoding in cortical structures and the role of reduced cholinergic modulation in allowing consolidation of previously encoded information. This article will focus on addressing the broad scope of different modulatory effects observed within hippocampal circuits, highlighting the heterogeneity of cholinergic modulation in terms of the physiological effects of activation of muscarinic and nicotinic receptors and the heterogeneity of effects on different subclasses of neurons.
Collapse
Affiliation(s)
- Holger Dannenberg
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, Boston, MA, United States
| | - Kimberly Young
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, Boston, MA, United States
| | - Michael Hasselmo
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, Boston, MA, United States
| |
Collapse
|
159
|
Gulyaeva NV, Bobkova NV, Kolosova NG, Samokhin AN, Stepanichev MY, Stefanova NA. Molecular and Cellular Mechanisms of Sporadic Alzheimer's Disease: Studies on Rodent Models in vivo. BIOCHEMISTRY (MOSCOW) 2017; 82:1088-1102. [PMID: 29037130 DOI: 10.1134/s0006297917100029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In this review, recent data are presented on molecular and cellular mechanisms of pathogenesis of the most widespread (about 95%) sporadic forms of Alzheimer's disease obtained on in vivo rodent models. Although none of the available models can fully reproduce the human disease, several key molecular mechanisms (such as dysfunction of neurotransmitter systems, especially of the acetylcholinergic system, β-amyloid toxicity, oxidative stress, neuroinflammation, mitochondrial dysfunction, disturbances in neurotrophic systems) are confirmed with different models. Injection models, olfactory bulbectomy, and senescence accelerated OXYS rats are reviewed in detail. These three approaches to in vivo modeling of sporadic Alzheimer's disease have demonstrated a considerable similarity in molecular and cellular mechanisms of pathology development. Studies on these models provide complementary data, and each model possesses its specific advantages. A general analysis of the data reported for the three models provides a multifaceted and the currently most complete molecular picture of sporadic Alzheimer's disease. This is highly relevant also from the practical viewpoint because it creates a basis for elaboration and preclinical studies of means for treatment of this disease.
Collapse
Affiliation(s)
- N V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia.
| | | | | | | | | | | |
Collapse
|
160
|
Grossberg S. Acetylcholine Neuromodulation in Normal and Abnormal Learning and Memory: Vigilance Control in Waking, Sleep, Autism, Amnesia and Alzheimer's Disease. Front Neural Circuits 2017; 11:82. [PMID: 29163063 PMCID: PMC5673653 DOI: 10.3389/fncir.2017.00082] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/12/2017] [Indexed: 01/30/2023] Open
Abstract
Adaptive Resonance Theory, or ART, is a neural model that explains how normal and abnormal brains may learn to categorize and recognize objects and events in a changing world, and how these learned categories may be remembered for a long time. This article uses ART to propose and unify the explanation of diverse data about normal and abnormal modulation of learning and memory by acetylcholine (ACh). In ART, vigilance control determines whether learned categories will be general and abstract, or specific and concrete. ART models how vigilance may be regulated by ACh release in layer 5 neocortical cells by influencing after-hyperpolarization (AHP) currents. This phasic ACh release is mediated by cells in the nucleus basalis (NB) of Meynert that are activated by unexpected events. The article additionally discusses data about ACh-mediated tonic control of vigilance. ART proposes that there are often dynamic breakdowns of tonic control in mental disorders such as autism, where vigilance remains high, and medial temporal amnesia, where vigilance remains low. Tonic control also occurs during sleep-wake cycles. Properties of Up and Down states during slow wave sleep arise in ACh-modulated laminar cortical ART circuits that carry out processes in awake individuals of contrast normalization, attentional modulation, decision-making, activity-dependent habituation, and mismatch-mediated reset. These slow wave sleep circuits interact with circuits that control circadian rhythms and memory consolidation. Tonic control properties also clarify how Alzheimer's disease symptoms follow from a massive structural degeneration that includes undermining vigilance control by ACh in cortical layers 3 and 5. Sleep disruptions before and during Alzheimer's disease, and how they contribute to a vicious cycle of plaque formation in layers 3 and 5, are also clarified from this perspective.
Collapse
Affiliation(s)
- Stephen Grossberg
- Center for Adaptive Systems, Graduate Program in Cognitive and Neural Systems, Departments of Mathematics & Statistics, Psychological & Brain Sciences and Biomedical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
161
|
Ke PC, Sani MA, Ding F, Kakinen A, Javed I, Separovic F, Davis TP, Mezzenga R. Implications of peptide assemblies in amyloid diseases. Chem Soc Rev 2017; 46:6492-6531. [PMID: 28702523 PMCID: PMC5902192 DOI: 10.1039/c7cs00372b] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neurodegenerative disorders and type 2 diabetes are global epidemics compromising the quality of life of millions worldwide, with profound social and economic implications. Despite the significant differences in pathology - much of which are poorly understood - these diseases are commonly characterized by the presence of cross-β amyloid fibrils as well as the loss of neuronal or pancreatic β-cells. In this review, we document research progress on the molecular and mesoscopic self-assembly of amyloid-beta, alpha synuclein, human islet amyloid polypeptide and prions, the peptides and proteins associated with Alzheimer's, Parkinson's, type 2 diabetes and prion diseases. In addition, we discuss the toxicities of these amyloid proteins based on their self-assembly as well as their interactions with membranes, metal ions, small molecules and engineered nanoparticles. Through this presentation we show the remarkable similarities and differences in the structural transitions of the amyloid proteins through primary and secondary nucleation, the common evolution from disordered monomers to alpha-helices and then to β-sheets when the proteins encounter the cell membrane, and, the consensus (with a few exceptions) that off-pathway oligomers, rather than amyloid fibrils, are the toxic species regardless of the pathogenic protein sequence or physicochemical properties. In addition, we highlight the crucial role of molecular self-assembly in eliciting the biological and pathological consequences of the amyloid proteins within the context of their cellular environments and their spreading between cells and organs. Exploiting such structure-function-toxicity relationship may prove pivotal for the detection and mitigation of amyloid diseases.
Collapse
Affiliation(s)
- Pu Chun Ke
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Marc-Antonie Sani
- School of Chemistry, Bio21 Institute, The University of Melbourne, 30 Flemington Rd, Parkville, VIC 3010, Australia
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Aleksandr Kakinen
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Ibrahim Javed
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Frances Separovic
- School of Chemistry, Bio21 Institute, The University of Melbourne, 30 Flemington Rd, Parkville, VIC 3010, Australia
| | - Thomas P. Davis
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Department of Chemistry, University of Warwick, Gibbet Hill, Coventry, CV4 7AL, United Kingdom
| | - Raffaele Mezzenga
- ETH Zurich, Department of Health Science & Technology, Schmelzbergstrasse 9, LFO, E23, 8092 Zurich, Switzerland
| |
Collapse
|
162
|
Brody AH, Strittmatter SM. Synaptotoxic Signaling by Amyloid Beta Oligomers in Alzheimer's Disease Through Prion Protein and mGluR5. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:293-323. [PMID: 29413525 PMCID: PMC5835229 DOI: 10.1016/bs.apha.2017.09.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) represents an impending global health crisis, yet the complexity of AD pathophysiology has so far precluded the development of any interventions to successfully slow or halt AD progression. It is clear that accumulation of Amyloid-beta (Aβ) peptide triggers progressive synapse loss to cause AD symptoms. Once initiated by Aβ, disease progression is complicated and accelerated by inflammation and by tau pathology. The recognition that Aβ peptide assumes multiple distinct states and that soluble oligomeric species (Aβo) are critical for synaptic damage is central to molecular understanding of AD. This knowledge has led to the identification of specific Aβo receptors, such as cellular prion protein (PrPC), mediating synaptic toxicity and neuronal dysfunction. The identification of PrPC as an Aβo receptor has illuminated an Aβo-induced signaling cascade involving mGluR5, Fyn, and Pyk2 that links Aβ and tau pathologies. This pathway provides novel potential therapeutic targets for disease-modifying AD therapy. Here, we discuss the methods by which several putative Aβo receptors were identified. We also offer an in-depth examination of the known molecular mechanisms believed to mediate Aβo-induced synaptic dysfunction, toxicity, and memory dysfunction.
Collapse
Affiliation(s)
- A Harrison Brody
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT, United States; Yale University, New Haven, CT, United States
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT, United States; Yale University, New Haven, CT, United States.
| |
Collapse
|
163
|
Brai E, Stuart S, Badin AS, Greenfield SA. A Novel Ex Vivo Model to Investigate the Underlying Mechanisms in Alzheimer's Disease. Front Cell Neurosci 2017; 11:291. [PMID: 29033787 PMCID: PMC5627036 DOI: 10.3389/fncel.2017.00291] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/04/2017] [Indexed: 01/09/2023] Open
Abstract
Currently there is no widely accepted animal model reproducing the full pathological profile of Alzheimer's disease (AD), since the basic mechanisms of neurodegeneration are still poorly understood. We have proposed that the interaction between the α7 nicotinic acetylcholine receptor (α7-nAChR) and a recently discovered toxic peptide, cleaved from the acetylcholinesterase (AChE) C-terminus, could account for the aberrant processes occurring in AD. In this article we describe a new application on ex vivo model procedure, which combines the advantages of both in vivo and in vitro preparations, to study the effects of the AChE-derived peptide on the rat basal forebrain (BF). Western blot analysis showed that the levels of α7-nAChR, p-Tau and Aβ are differentially expressed upon the AChE-peptide administration, in a selective site-dependent manner. In conclusion, this methodology demonstrates the action of a novel peptide in triggering an AD-like phenotype and proposes a new ex vivo approach for manipulating and monitoring neurochemical processes contributing to neurodegeneration, in a time-dependent and site-specific manner.
Collapse
Affiliation(s)
- Emanuele Brai
- Neuro-Bio Ltd., Culham Science CentreAbingdon, United Kingdom
| | - Skye Stuart
- Neuro-Bio Ltd., Culham Science CentreAbingdon, United Kingdom.,School of Physiology, Pharmacology and Neuroscience, Faculty of Biomedical Sciences, University of BristolBristol, United Kingdom
| | | | | |
Collapse
|
164
|
BACE1 Function and Inhibition: Implications of Intervention in the Amyloid Pathway of Alzheimer's Disease Pathology. Molecules 2017; 22:molecules22101723. [PMID: 29027981 PMCID: PMC6151801 DOI: 10.3390/molecules22101723] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a fatal progressive neurodegenerative disorder characterized by increasing loss in memory, cognition, and function of daily living. Among the many pathologic events observed in the progression of AD, changes in amyloid β peptide (Aβ) metabolism proceed fastest, and precede clinical symptoms. BACE1 (β-secretase 1) catalyzes the initial cleavage of the amyloid precursor protein to generate Aβ. Therefore inhibition of BACE1 activity could block one of the earliest pathologic events in AD. However, therapeutic BACE1 inhibition to block Aβ production may need to be balanced with possible effects that might result from diminished physiologic functions BACE1, in particular processing of substrates involved in neuronal function of the brain and periphery. Potentials for beneficial or consequential effects resulting from pharmacologic inhibition of BACE1 are reviewed in context of ongoing clinical trials testing the effect of BACE1 candidate inhibitor drugs in AD populations.
Collapse
|
165
|
Cartereau A, Martin C, Thany SH. Neonicotinoid insecticides differently modulate acetycholine-induced currents on mammalian α7 nicotinic acetylcholine receptors. Br J Pharmacol 2017; 175:1987-1998. [PMID: 28853147 DOI: 10.1111/bph.14018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 08/11/2017] [Accepted: 08/17/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Neonicotinoid insecticides are described as poor agonists of mammalian nicotinic ACh receptors. In this paper, we show that their effects on mammalian nicotinic receptors differ between compounds. EXPERIMENTAL APPROACH Two-electrode voltage-clamp electrophysiology was used to characterize the pharmacology of three neonicotinoid insecticides on nicotinic α7 receptors expressed in Xenopus oocytes. Single and combined application of clothianidin, acetamiprid and thiamethoxam were tested. RESULTS Two neonicotinoid insecticides, clothianidin and acetamiprid, were partial agonists of mammalian neuronal α7 nicotinic receptors, whereas another neonicotinoid insecticide, thiamethoxam, which is converted to clothianidin in insect and plant tissues, had no effect. Pretreatment with clothianidin and acetamiprid (10 μM) ACh significantly enhanced the subsequent currents evoked by ACh (100 μM ) whereas pretreatment with thiamethoxam (10 μM) reduced ACh-induced current amplitudes.A combination of the three neonicotinoids decreased the ACh-evoked currents. CONCLUSIONS AND IMPLICATIONS The present findings suggest that neonicotinoid insecticides differ markedly in their direct effects on mammalian α7 nicotinic ACh receptors and can also modulate ACh-induced currents. Furthermore, our data indicate a previously unknown modulation of mammalian α7 nicotinic receptors by a combination of clothianidin, acetamiprid and thiamethoxam. LINKED ARTICLES This article is part of a themed section on Nicotinic Acetylcholine Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.11/issuetoc.
Collapse
Affiliation(s)
- Alison Cartereau
- LBLGC, UPRES EA 1207-USC INRA 1328, Université d'Orléans, Orléans, France
| | - Carine Martin
- LBLGC, UPRES EA 1207-USC INRA 1328, Université d'Orléans, Orléans, France
| | - Steeve H Thany
- LBLGC, UPRES EA 1207-USC INRA 1328, Université d'Orléans, Orléans, France
| |
Collapse
|
166
|
Ablating ErbB4 in PV neurons attenuates synaptic and cognitive deficits in an animal model of Alzheimer's disease. Neurobiol Dis 2017; 106:171-180. [DOI: 10.1016/j.nbd.2017.07.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/16/2017] [Accepted: 07/02/2017] [Indexed: 02/07/2023] Open
|
167
|
Lykhmus O, Uspenska K, Koval L, Lytovchenko D, Voytenko L, Horid'ko T, Kosiakova H, Gula N, Komisarenko S, Skok M. N-Stearoylethanolamine protects the brain and improves memory of mice treated with lipopolysaccharide or immunized with the extracellular domain of α7 nicotinic acetylcholine receptor. Int Immunopharmacol 2017; 52:290-296. [PMID: 28963942 DOI: 10.1016/j.intimp.2017.09.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/12/2017] [Accepted: 09/25/2017] [Indexed: 12/11/2022]
Abstract
Neuroinflammation is an important risk factor for neurodegenerative disorders like Alzheimer's disease. Nicotinic acetylcholine receptors of α7 subtype (α7 nAChRs) regulate inflammatory processes in various tissues, including the brain. N-stearoylethanolamine (NSE) is a biologically active cell membrane component with anti-inflammatory and membrane-protective properties. Previously we found that mice injected with bacterial lipopolysaccharide (LPS) or immunized with recombinant extracellular domain (1-208) of α7 nAChR subunit possessed decreased α7 nAChR levels, accumulated pathogenic amyloid-beta peptide Aβ(1-42) in the brain and demonstrated impaired episodic memory compared to non-treated mice. Here we studied the effect of NSE on behavior and brain components of LPS- treated or α7(1-208)-immunized mice. NSE, given per os, non-significantly decreased LPS-stimulated interleukin-6 elevation in the brain, slowed down the α7(1-208)-specific IgG antibody production and prevented the antibody penetration into the brain of mice. NSE prevented the loss of α7 nAChRs and accumulation of α7-bound Aβ(1-42) in the brain and brain mitochondria of LPS-treated or α7(1-208)-immunized mice and supported mitochondria resistance to apoptosis by attenuating Ca2+-stimulated cytochrome c release. Finally, NSE significantly improved episodic memory of mice impaired by either LPS treatment or immunization with α7(1-208). The results of our study demonstrate a therapeutic potential of NSE for prevention of cognitive disfunction caused by neuroinflammation or autoimmune reaction that allows suggesting this drug as a candidate for the treatment or prophylaxis of Alzheimer's pathology.
Collapse
Affiliation(s)
- Olena Lykhmus
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry, 9, Leontovycha str, 01030 Kyiv, Ukraine
| | - Kateryna Uspenska
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry, 9, Leontovycha str, 01030 Kyiv, Ukraine
| | - Lyudmyla Koval
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry, 9, Leontovycha str, 01030 Kyiv, Ukraine
| | - Daria Lytovchenko
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry, 9, Leontovycha str, 01030 Kyiv, Ukraine
| | - Larysa Voytenko
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry, 9, Leontovycha str, 01030 Kyiv, Ukraine
| | - Tetyana Horid'ko
- Department of Lipid Biochemistry, Palladin Institute of Biochemistry, 9, Leontovycha str, 01030 Kyiv, Ukraine
| | - Halyna Kosiakova
- Department of Lipid Biochemistry, Palladin Institute of Biochemistry, 9, Leontovycha str, 01030 Kyiv, Ukraine
| | - Nadiya Gula
- Department of Lipid Biochemistry, Palladin Institute of Biochemistry, 9, Leontovycha str, 01030 Kyiv, Ukraine
| | - Serhiy Komisarenko
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry, 9, Leontovycha str, 01030 Kyiv, Ukraine
| | - Maryna Skok
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry, 9, Leontovycha str, 01030 Kyiv, Ukraine.
| |
Collapse
|
168
|
Wang HY, Trocmé-Thibierge C, Stucky A, Shah SM, Kvasic J, Khan A, Morain P, Guignot I, Bouguen E, Deschet K, Pueyo M, Mocaer E, Ousset PJ, Vellas B, Kiyasova V. Increased Aβ 42-α7-like nicotinic acetylcholine receptor complex level in lymphocytes is associated with apolipoprotein E4-driven Alzheimer's disease pathogenesis. ALZHEIMERS RESEARCH & THERAPY 2017; 9:54. [PMID: 28750690 PMCID: PMC5530996 DOI: 10.1186/s13195-017-0280-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/21/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND The apolipoprotein E ε4 (APOE4) genotype is a prominent late-onset Alzheimer's disease (AD) risk factor. ApoE4 disrupts memory function in rodents and may contribute to both plaque and tangle formation. METHODS Coimmunoprecipitation and Western blot detection were used to determine: 1) the effects of select fragments from the apoE low-density lipoprotein (LDL) binding domain and recombinant apoE subtypes on amyloid beta (Aβ)42-α7 nicotinic acetylcholine receptor (α7nAChR) interaction and tau phosphorylation in rodent brain synaptosomes; and 2) the level of Aβ42-α7nAChR complexes in matched controls and patients with mild cognitive impairment (MCI) and dementia due to AD with known APOE genotypes. RESULTS In an ex vivo study using rodent synaptosomes, apoE141-148 of the apoE promotes Aβ42-α7nAChR association and Aβ42-induced α7nAChR-dependent tau phosphorylation. In a single-blind study, we examined lymphocytes isolated from control subjects, patients with MCI and dementia due to AD with known APOE genotypes, sampled at two time points (1 year apart). APOE ε4 genotype was closely correlated with heightened Aβ42-α7nAChR complex levels and with blunted exogenous Aβ42 effects in lymphocytes derived from AD and MCI due to AD cases. Similarly, plasma from APOE ε4 carriers enhanced the Aβ42-induced Aβ42-α7nAChR association in rat cortical synaptosomes. The progression of cognitive decline in APOE ε4 carriers correlated with higher levels of Aβ42-α7nAChR complexes in lymphocytes and greater enhancement by their plasma of Aβ42-induced Aβ42-α7nAChR association in rat cortical synaptosomes. CONCLUSIONS Our data suggest that increased lymphocyte Aβ42-α7nAChR-like complexes may indicate the presence of AD pathology especially in APOE ε4 carriers. We show that apoE, especially apoE4, promotes Aβ42-α7nAChR interaction and Aβ42-induced α7nAChR-dependent tau phosphorylation via its apoE141-148 domain. These apoE-mediated effects may contribute to the APOE ε4-driven neurodysfunction and AD pathologies.
Collapse
Affiliation(s)
- Hoau-Yan Wang
- Department of Physiology, Pharmacology and Neuroscience, CUNY School of Medicine, 160 Convent Avenue, New York, NY, 10031, USA. .,Department of Physiology, Pharmacology & Neuroscience, The City University of New York School of Medicine, CDI-3370 85 St. Nicholas Terrace, New York, NY, 10027, USA.
| | | | - Andres Stucky
- Department of Physiology, Pharmacology and Neuroscience, CUNY School of Medicine, 160 Convent Avenue, New York, NY, 10031, USA.,Department of Biology, Neuroscience Program, Graduate School of The City University of New York, New York, New York, 10061, USA
| | - Sanket M Shah
- Department of Physiology, Pharmacology and Neuroscience, CUNY School of Medicine, 160 Convent Avenue, New York, NY, 10031, USA
| | - Jessica Kvasic
- Department of Physiology, Pharmacology and Neuroscience, CUNY School of Medicine, 160 Convent Avenue, New York, NY, 10031, USA
| | - Amber Khan
- Department of Physiology, Pharmacology and Neuroscience, CUNY School of Medicine, 160 Convent Avenue, New York, NY, 10031, USA
| | - Philippe Morain
- Institut de Recherches Internationales Servier, 50 Rue Carnot, 92284, Suresnes, France
| | - Isabelle Guignot
- Institut de Recherches Internationales Servier, 50 Rue Carnot, 92284, Suresnes, France
| | - Eva Bouguen
- Institut de Recherches Internationales Servier, 50 Rue Carnot, 92284, Suresnes, France
| | - Karine Deschet
- Institut de Recherches Internationales Servier, 50 Rue Carnot, 92284, Suresnes, France
| | - Maria Pueyo
- Institut de Recherches Internationales Servier, 50 Rue Carnot, 92284, Suresnes, France
| | - Elisabeth Mocaer
- Institut de Recherches Internationales Servier, 50 Rue Carnot, 92284, Suresnes, France
| | - Pierre-Jean Ousset
- Alzheimer's Disease Research and Clinical Center, Inserm U1027, Toulouse University Hospital, Toulouse, France
| | - Bruno Vellas
- Alzheimer's Disease Research and Clinical Center, Inserm U1027, Toulouse University Hospital, Toulouse, France
| | - Vera Kiyasova
- Institut de Recherches Internationales Servier, 50 Rue Carnot, 92284, Suresnes, France
| |
Collapse
|
169
|
Lazarevic V, Fieńko S, Andres-Alonso M, Anni D, Ivanova D, Montenegro-Venegas C, Gundelfinger ED, Cousin MA, Fejtova A. Physiological Concentrations of Amyloid Beta Regulate Recycling of Synaptic Vesicles via Alpha7 Acetylcholine Receptor and CDK5/Calcineurin Signaling. Front Mol Neurosci 2017; 10:221. [PMID: 28785201 PMCID: PMC5520466 DOI: 10.3389/fnmol.2017.00221] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/26/2017] [Indexed: 01/09/2023] Open
Abstract
Despite the central role of amyloid β (Aβ) peptide in the etiopathogenesis of Alzheimer’s disease (AD), its physiological function in healthy brain is still debated. It is well established that elevated levels of Aβ induce synaptic depression and dismantling, connected with neurotoxicity and neuronal loss. Growing evidence suggests a positive regulatory effect of Aβ on synaptic function and cognition; however the exact cellular and molecular correlates are still unclear. In this work, we tested the effect of physiological concentrations of Aβ species of endogenous origin on neurotransmitter release in rat cortical and hippocampal neurons grown in dissociated cultures. Modulation of production and degradation of the endogenous Aβ species as well as applications of the synthetic rodent Aβ40 and Aβ42 affected efficacy of neurotransmitter release from individual presynapses. Low picomolar Aβ40 and Aβ42 increased, while Aβ depletion or application of low micromolar concentration decreased synaptic vesicle recycling, showing a hormetic effect of Aβ on neurotransmitter release. These Aβ-mediated modulations required functional alpha7 acetylcholine receptors as well as extracellular and intracellular calcium, involved regulation of CDK5 and calcineurin signaling and increased recycling of synaptic vesicles. These data indicate that Aβ regulates neurotransmitter release from presynapse and suggest that failure of the normal physiological function of Aβ in the fine-tuning of SV cycling could disrupt synaptic function and homeostasis, which would, eventually, lead to cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Vesna Lazarevic
- RG Presynaptic Plasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany.,Department of Neurochemistry and Molecular Biology, Leibniz Institute for NeurobiologyMagdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE)Magdeburg, Germany
| | - Sandra Fieńko
- RG Presynaptic Plasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Maria Andres-Alonso
- RG Presynaptic Plasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Daniela Anni
- Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Hospital, University of Erlangen-NurembergErlangen, Germany
| | - Daniela Ivanova
- RG Presynaptic Plasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | | | - Eckart D Gundelfinger
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for NeurobiologyMagdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE)Magdeburg, Germany.,Center for Behavioral Brain Sciences, Otto von Guericke UniversityMagdeburg, Germany.,Medical Faculty, Otto von Guericke UniversityMagdeburg, Germany
| | - Michael A Cousin
- Centre for Integrative Physiology, University of EdinburghEdinburgh, United Kingdom
| | - Anna Fejtova
- RG Presynaptic Plasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany.,Department of Neurochemistry and Molecular Biology, Leibniz Institute for NeurobiologyMagdeburg, Germany.,Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Hospital, University of Erlangen-NurembergErlangen, Germany.,Center for Behavioral Brain Sciences, Otto von Guericke UniversityMagdeburg, Germany
| |
Collapse
|
170
|
Cruz-Haces M, Tang J, Acosta G, Fernandez J, Shi R. Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases. Transl Neurodegener 2017; 6:20. [PMID: 28702179 PMCID: PMC5504572 DOI: 10.1186/s40035-017-0088-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury is among the most common causes of death and disability in youth and young adults. In addition to the acute risk of morbidity with moderate to severe injuries, traumatic brain injury is associated with a number of chronic neurological and neuropsychiatric sequelae including neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. However, despite the high incidence of traumatic brain injuries and the established clinical correlation with neurodegeneration, the causative factors linking these processes have not yet been fully elucidated. Apart from removal from activity, few, if any prophylactic treatments against post-traumatic brain injury neurodegeneration exist. Therefore, it is imperative to understand the pathophysiological mechanisms of traumatic brain injury and neurodegeneration in order to identify potential factors that initiate neurodegenerative processes. Oxidative stress, neuroinflammation, and glutamatergic excitotoxicity have previously been implicated in both secondary brain injury and neurodegeneration. In particular, reactive oxygen species appear to be key in mediating molecular insult in neuroinflammation and excitotoxicity. As such, it is likely that post injury oxidative stress is a key mechanism which links traumatic brain injury to increased risk of neurodegeneration. Consequently, reactive oxygen species and their subsequent byproducts may serve as novel fluid markers for identification and monitoring of cellular damage. Furthermore, these reactive species may further serve as a suitable therapeutic target to reduce the risk of post-injury neurodegeneration and provide long term quality of life improvements for those suffering from traumatic brain injury.
Collapse
Affiliation(s)
- Marcela Cruz-Haces
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Jonathan Tang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Glen Acosta
- Department of Basic Medical Sciences, Purdue University, West Lafayette, USA
| | - Joseph Fernandez
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Riyi Shi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
- Department of Basic Medical Sciences, Purdue University, West Lafayette, USA
| |
Collapse
|
171
|
Wang HY, Lee KC, Pei Z, Khan A, Bakshi K, Burns LH. PTI-125 binds and reverses an altered conformation of filamin A to reduce Alzheimer's disease pathogenesis. Neurobiol Aging 2017; 55:99-114. [DOI: 10.1016/j.neurobiolaging.2017.03.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/06/2017] [Accepted: 03/09/2017] [Indexed: 11/30/2022]
|
172
|
Shrivastava AN, Aperia A, Melki R, Triller A. Physico-Pathologic Mechanisms Involved in Neurodegeneration: Misfolded Protein-Plasma Membrane Interactions. Neuron 2017; 95:33-50. [DOI: 10.1016/j.neuron.2017.05.026] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 05/12/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
|
173
|
Lagarde J, Sarazin M, Bottlaender M. In vivo PET imaging of neuroinflammation in Alzheimer's disease. J Neural Transm (Vienna) 2017; 125:847-867. [PMID: 28516240 DOI: 10.1007/s00702-017-1731-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/01/2017] [Indexed: 12/15/2022]
Abstract
Increasing evidence suggests that neuroinflammation contributes to the pathophysiology of many neurodegenerative diseases, especially Alzheimer's disease (AD). Molecular imaging by PET may be a useful tool to assess neuroinflammation in vivo, thus helping to decipher the complex role of inflammatory processes in the pathophysiology of neurodegenerative diseases and providing a potential means of monitoring the effect of new therapeutic approaches. For this objective, the main target of PET studies is the 18 kDa translocator protein (TSPO), as it is overexpressed by activated microglia. In the present review, we describe the most widely used PET tracers targeting the TSPO, the methodological issues in tracer quantification and summarize the results obtained by TSPO PET imaging in AD, as well as in neurodegenerative disorders associated with AD, in psychiatric disorders and ageing. We also briefly describe alternative PET targets and imaging modalities to study neuroinflammation. Lastly, we question the meaning of PET imaging data in the context of a highly complex and multifaceted role of neuroinflammation in neurodegenerative diseases. This overview leads to the conclusion that PET imaging of neuroinflammation is a promising way of deciphering the enigma of the pathophysiology of AD and of monitoring the effect of new therapies.
Collapse
Affiliation(s)
- Julien Lagarde
- Unit of Neurology of Memory and Language, Centre de Psychiatrie et Neurosciences, INSERM UMR S894, Centre Hospitalier Sainte-Anne and Université Paris Descartes, Sorbonne Paris Cité, 75014, Paris, France
| | - Marie Sarazin
- Unit of Neurology of Memory and Language, Centre de Psychiatrie et Neurosciences, INSERM UMR S894, Centre Hospitalier Sainte-Anne and Université Paris Descartes, Sorbonne Paris Cité, 75014, Paris, France
| | - Michel Bottlaender
- UNIACT, NeuroSpin, Institut d'Imagerie Biomédicale, Direction de la Recherche Fondamentale, Commissariat à l'Energie Atomique, 91191, Gif-sur-Yvette, France. .,Laboratoire Imagerie Moléculaire in Vivo, UMR 1023, Service Hospitalier Frédéric Joliot, Institut d'Imagerie Biomédicale, Direction de la Recherche Fondamentale, Commissariat à l'Energie Atomique, 91400, Orsay, France.
| |
Collapse
|
174
|
Ekins S, Mathews P, Saito EK, Diaz N, Naylor D, Chung J, McMurtray AM. α7-Nicotinic acetylcholine receptor inhibition by indinavir: implications for cognitive dysfunction in treated HIV disease. AIDS 2017; 31:1083-1089. [PMID: 28358738 DOI: 10.1097/qad.0000000000001488] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The study set out to determine if the HIV protease inhibitor, indinavir, alters responsiveness of α7-nicotinic acetylcholine receptors to acetylcholine. DESIGN Treatment with HAART has dramatically reduced development of HIV-associated dementia and more severe forms of cognitive impairment. However, many individuals continue to experience cognitive decline of uncertain cause. Previous studies have failed to demonstrate significant alterations of functional brain connectivity, structural brain changes, or changes in cerebral blood flow sufficient to explain cognitive decline in virally suppressed individuals. This suggests that the mechanisms underlying development and progression of cognitive problems likely occurs at a micro rather than macro level, such as disruptions in neurotransmitter system signaling. MATERIALS AND METHODS Indinavir's effects on α7-nicotinic acetylcholine receptor activity was tested using a ScreenPatch IonWorks Barracuda-based assay in a mammalian cell model. RESULTS At low concentrations (0.0003-10 μmol/l) indinavir acts as a positive allosteric modulator (EC50 = 0.021 μmol/l), whereas at concentrations greater than 10 μmol/l (30-100 μmol/l) indinavir acts as an inhibitor of the α7-nicotinic acetylcholine receptor. CONCLUSION At concentrations greater than 10 μmol/l indinavir reduces synaptic transmission in the acetylcholine neurotransmitter system, which could possibly contribute to cognitive dysfunction. These results suggest that further experiments should be considered to assess whether patients might benefit from treatment with cholinesterase inhibitors that counteract the effects of indinavir.
Collapse
|
175
|
Bao W, Jia H, Finnema S, Cai Z, Carson RE, Huang YH. PET Imaging for Early Detection of Alzheimer's Disease: From Pathologic to Physiologic Biomarkers. PET Clin 2017; 12:329-350. [PMID: 28576171 DOI: 10.1016/j.cpet.2017.03.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This article describes the application of various PET imaging agents in the investigation and diagnosis of Alzheimer's disease (AD), including radiotracers for pathologic biomarkers of AD such as β-amyloid deposits and tau protein aggregates, and the neuroinflammation biomarker 18 kDa translocator protein, as well as physiologic biomarkers, such as cholinergic receptors, glucose metabolism, and the synaptic density biomarker synaptic vesicle glycoprotein 2A. Potential of these biomarkers for early AD diagnosis is also assessed.
Collapse
Affiliation(s)
- Weiqi Bao
- PET Center, Huanshan Hospital, Fudan University, No. 518, East Wuzhong Road, Xuhui District, Shanghai 200235, China
| | - Hongmei Jia
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, No. 19, Xinjiekouwai Street, Beijing 10075, China
| | - Sjoerd Finnema
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA
| | - Yiyun Henry Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, PO Box 208048, New Haven, CT 06520-8048, USA.
| |
Collapse
|
176
|
Kim HY, Kwon JA, Kang T, Choi I. Rapid and high-throughput colorimetric screening for anti-aggregation reagents of protein conformational diseases by using gold nanoplasmonic particles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1575-1585. [DOI: 10.1016/j.nano.2017.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/29/2016] [Accepted: 01/09/2017] [Indexed: 01/04/2023]
|
177
|
Melki R. How the shapes of seeds can influence pathology. Neurobiol Dis 2017; 109:201-208. [PMID: 28363800 DOI: 10.1016/j.nbd.2017.03.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/16/2017] [Accepted: 03/26/2017] [Indexed: 10/19/2022] Open
Abstract
It is widely accepted that the loss of function of different cellular proteins following their aggregation into highly stable aggregates or the gain of pathologic function of the resulting macromolecular assemblies or both processes are tightly associated to distinct debilitating neurodegenerative diseases such as Alzheimer's, Parkinson's, Creutzfeldt-Jacob, Amyotrophic Lateral Sclerosis and Huntington's diseases. How the aggregation of one given protein leads to distinct diseases is unclear. Here, a structural-molecular explanation based on the ability of proteins such as α-synuclein or tau to form assemblies that differ by their intrinsic architecture, stability, seeding capacity, and surfaces is proposed to account for distinct synucleinopathies and tauopathies. The shape and surfaces of the seeds is proposed to define at the same time their seeding capacity, interactome and tropism for defined neuronal cells within the central nervous system.
Collapse
Affiliation(s)
- Ronald Melki
- Paris Saclay Institute of Neurosciences, CNRS, Bâtiment 32-33, 1 Avenue de la Terrasse, 91190 Gif-sur-Yvette, France.
| |
Collapse
|
178
|
Hisatsune C, Mikoshiba K. IP 3 receptor mutations and brain diseases in human and rodents. J Neurochem 2017; 141:790-807. [PMID: 28211945 DOI: 10.1111/jnc.13991] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/03/2017] [Accepted: 02/12/2017] [Indexed: 01/15/2023]
Abstract
The inositol 1,4,5-trisphosphate receptor (IP3 R) is a huge Ca2+ channel that is localized at the endoplasmic reticulum. The IP3 R releases Ca2+ from the endoplasmic reticulum upon binding to IP3 , which is produced by various extracellular stimuli through phospholipase C activation. All vertebrate organisms have three subtypes of IP3 R genes, which have distinct properties of IP3 -binding and Ca2+ sensitivity, and are differently regulated by phosphorylation and by their associated proteins. Each cell type expresses the three subtypes of IP3 R in a distinct proportion, which is important for creating and maintaining spatially and temporally appropriate intracellular Ca2+ level patterns for the regulation of specific physiological phenomena. Of the three types of IP3 Rs, the type 1 receptor (IP3 R1) is dominantly expressed in the brain and is important for brain function. Recent emerging evidence suggests that abnormal Ca2+ signals from the IP3 R1 are closely associated with human brain pathology. In this review, we focus on the recent advances in our knowledge of the regulation of IP3 R1 and its functional implication in human brain diseases, as revealed by IP3 R mutation studies and analysis of human disease-associated genes. This article is part of the mini review series "60th Anniversary of the Japanese Society for Neurochemistry".
Collapse
Affiliation(s)
- Chihiro Hisatsune
- Laboratory for Developmental Neurobiology, Brain Science Institute, Institute of Physical and Chemical Research (RIKEN), Saitama, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, Brain Science Institute, Institute of Physical and Chemical Research (RIKEN), Saitama, Japan
| |
Collapse
|
179
|
Vicens P, Heredia L, Torrente M, Domingo JL. Behavioural effects of PNU-282987 and stress in an animal model of Alzheimer's disease. Psychogeriatrics 2017; 17:33-42. [PMID: 26817787 DOI: 10.1111/psyg.12189] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/11/2015] [Accepted: 12/17/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cholinergic deficits play an important role in both cognitive and behavioural alterations in Alzheimer's disease. This study was aimed at evaluating the possible therapeutic role of PNU-282987 (PNU), an α7 nicotinic cholinergic receptor agonist, and the possible effects of stress in precipitating the onset of behavioural deficits in animals with susceptibility to Alzheimer's disease. METHODS B6C3-Tg mice with susceptibility to Alzheimer's disease and wild-type mice either with or without restraint stress received 0- or 1-mg/kg PNU. At 12 months old, mice were evaluated for activity levels, anxiety-like levels, and spatial learning and memory. RESULTS Data did not show the effects of PNU on activity and anxiety-like behaviour. No effect of PNU on acquisition of a spatial learning task was detected, but a reversal of stress effects on retention in the Morris water maze was observed in transgenic mice. CONCLUSIONS Further studies are needed in order to better understand the role of α7 nicotinic cholinergic receptor agonists in motor activity, anxiety, and spatial learning and memory and to develop more accurate pharmacological treatment of psychopathological diseases.
Collapse
Affiliation(s)
- Paloma Vicens
- Department of Psychology, Universitat Rovira i Virgili, Tarragona, Spain.,Research Center in Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain.,Laboratory of Toxicology and Environmental Health, School of Medicine, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona, Spain
| | - Luis Heredia
- Department of Psychology, Universitat Rovira i Virgili, Tarragona, Spain.,Research Center in Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain.,Laboratory of Toxicology and Environmental Health, School of Medicine, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona, Spain
| | - Margarita Torrente
- Department of Psychology, Universitat Rovira i Virgili, Tarragona, Spain.,Research Center in Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain.,Laboratory of Toxicology and Environmental Health, School of Medicine, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona, Spain
| | - José L Domingo
- Laboratory of Toxicology and Environmental Health, School of Medicine, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona, Spain
| |
Collapse
|
180
|
Burns LH, Wang HY. Altered filamin A enables amyloid beta-induced tau hyperphosphorylation and neuroinflammation in Alzheimer's disease. NEUROIMMUNOLOGY AND NEUROINFLAMMATION 2017; 4:263-271. [PMID: 34295950 PMCID: PMC8294116 DOI: 10.20517/2347-8659.2017.50] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease with proteopathy characterized by abnormalities in amyloid beta (Aβ) and tau proteins. Defective amyloid and tau propagate and aggregate, leading to eventual amyloid plaques and neurofibrillary tangles. New data show that a third proteopathy, an altered conformation of the scaffolding protein filamin A (FLNA), is critically linked to the amyloid and tau pathologies in AD. Altered FLNA is pervasive in AD brain and without apparent aggregation. In a striking interdependence, altered FLNA is both induced by Aβ and required for two prominent pathogenic signaling pathways of Aβ. Aβ monomers or small oligomers signal via the α7 nicotinic acetylcholine receptor (α7nAChR) to activate kinases that hyperphosphorylate tau to cause neurofibrillary lesions and formation of neurofibrillary tangles. Altered FLNA also enables a persistent activation of toll-like-receptor 4 (TLR4) by Aβ, leading to excessive inflammatory cytokine release and neuroinflammation. The novel AD therapeutic candidate PTI-125 binds and reverses the altered FLNA conformation to prevent Aβ’s signaling via α7nAChR and aberrant activation of TLR4, thus reducing multiple AD-related neuropathologies. As a regulator of Aβ’s signaling via α7nAChR and TLR4, altered FLNA represents a novel AD therapeutic target.
Collapse
Affiliation(s)
| | - Hoau-Yan Wang
- Department of Physiology, Pharmacology and Neuroscience, City University of New York School of Medicine, New York, NY 10031, USA.,Department of Biology and Neuroscience, Graduate School of the City University of New York, New York, NY 10031, USA
| |
Collapse
|
181
|
Alzheimer's Disease: Insights from Genetic Mouse Models and Current Advances in Human IPSC-Derived Neurons. ADVANCES IN NEUROBIOLOGY 2017; 15:3-29. [PMID: 28674976 DOI: 10.1007/978-3-319-57193-5_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease was first described in 1906 and since then tremendous efforts have been made to fully understand the disease pathology and to find a cure for this neurodegenerative disease. The diagnosis of Alzheimer's is still difficult, especially in early stages of the disease. Current treatment of Alzheimer's only ameliorates the symptoms but fails to provide a therapy. Over the last decades, animal models have been proven valuable in elucidating insights of the pathology. In vitro models using patient-derived cells are currently emerging and hold great promise in understanding the disease pathophysiology. Here, we introduce the neurobiology and genetic features of Alzheimer's and describe what we have learned from studies employing mouse models and patient-derived induced pluripotent stem cells.
Collapse
|
182
|
Ahmed T, Zahid S, Mahboob A, Farhat SM. Cholinergic System and Post-translational Modifications: An Insight on the Role in Alzheimer's Disease. Curr Neuropharmacol 2017; 15:480-494. [PMID: 27012953 PMCID: PMC5543671 DOI: 10.2174/1570159x14666160325121145] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/02/2015] [Accepted: 03/03/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common form of old age dementia. The formation of amyloid plaques (Aβ), neurofibrillary tangles and loss of basal forebrain cholinergic neurons are the hallmark events in the pathology of AD. LITERATURE REVIEW Cholinergic system is one of the most important neurotransmitter system involved in learning and memory which preferentially degenerates in the initial stages of AD. Activation of cholinergic receptors (muscarinic and nicotinic) activates multiple pathways which result in post translational modifications (PTMs) in multiple proteins which bring changes in nervous system. Cholinergic receptors-mediated PTMs "in-part" substantially affect the biosynthesis, proteolysis, degradation and expression of many proteins and in particular, amyloid precursor protein (APP). APP is subjected to several PTMs (proteolytic processing, glycosylation, sulfation, and phosphorylation) during its course of processing, resulting in Aβ deposition, leading to AD. Aβ also alters the PTMs of tau which is a microtubule associated protein. Therefore, post-translationally modified tau and Aβ collectively aggravate the neuronal loss that leads to cholinergic hypofunction. CONCLUSION Despite the accumulating evidences, the interaction between cholinergic neurotransmission and the physiological significance of PTM events remain speculative and still needs further exploration. This review focuses on the role of cholinergic system and discusses the significance of PTMs in pathological progression of AD and highlights some important future directions.
Collapse
Affiliation(s)
- Touqeer Ahmed
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan
| | - Saadia Zahid
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan
| | | | | |
Collapse
|
183
|
Paulo JA, Gygi SP. Nicotine-induced protein expression profiling reveals mutually altered proteins across four human cell lines. Proteomics 2016; 17. [PMID: 27862958 DOI: 10.1002/pmic.201600319] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/11/2016] [Accepted: 11/10/2016] [Indexed: 11/10/2022]
Abstract
Mass spectrometry-based proteomic strategies can profile the expression level of proteins in response to external stimuli. Nicotine affects diverse cellular pathways, however, the nicotine-induced alterations on the global proteome across human cell lines have not been fully elucidated. We measured perturbations in protein levels resulting from nicotine treatment in four cell lines-HEK, HeLa, PaSC, and SH-SY5Y-in a single experiment using tandem mass tags (TMT10-plex) and high-resolution mass spectrometry. We quantified 8590 proteins across all cell lines. Of these, nicotine increased the abundance of 31 proteins 1.5-fold or greater in all cell lines. Likewise, considering proteins with altered levels in at least three of the four cell lines, 64 were up-regulated, while one was down-regulated. Gene ontology analysis revealed that ∼40% of these proteins were membrane bound, and functioned in transmembrane signaling and receptor activity. We highlighted proteins, including APP, APLP2, LAPTM4B, and NCOA4, which were dysregulated by nicotine in all cell lines investigated and may have implications in downstream signaling pathways, particularly autophagy. Using the outlined methodology, studies in additional (including primary) cell lines will provide further evidence that alterations in the levels of these proteins are indeed a general response to nicotine and thereby merit further investigation.
Collapse
Affiliation(s)
- Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
184
|
Gupta A, Goyal R. Amyloid beta plaque: a culprit for neurodegeneration. Acta Neurol Belg 2016; 116:445-450. [PMID: 27118573 DOI: 10.1007/s13760-016-0639-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/04/2016] [Indexed: 01/08/2023]
Abstract
Increasing life expectancy has resulted in an increase in neurodegenerative disorders like Alzheimer's disease. None of the hypothesis proposed till date explains the exact pathobiology of the disease. It is therefore imperative to understand the underlying mechanisms. Amyloid beta (Aβ) is regarded as the main culprit and maximum therapeutic efforts are centered towards Aβ. This review will discuss about the biosynthesis, the physiological role of Aβ including the pathogenic aggregation of Aβ resulting neurodegenerative cognitive disabilities. Most studies of Alzheimer's disease have focused on the biochemical mechanisms involved in the neurodegenerative processes triggered by Aβ aggregates. Aβ is generated from mature amyloid precursor protein being metabolized by two competing pathways, α-secretase pathway (non-amyloidogenic pathway) and β-secretase (amyloidogenic pathway). The physiological roles of Aβ reported in neurotrophic properties, neurogenesis, synaptic plasticity, metal ion sequestration and specificity of blood brain barrier. The neuronal injury is the result of Aβ oligomerization and it is reported that oligomerization of Aβ contributes to neurodegeneration in Alzheimer's disease. The physiological role of Aβ must be considered in the development of medications that intended to decrease its oligomerization forming plaques in a disease like Alzheimer's disease. The biosynthetic pathways for transport and accumulation of Aβ need to be ascertained as an attempt to develop future strategies for prevention of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ankita Gupta
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173212, India
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173212, India.
| |
Collapse
|
185
|
Kam TI, Park H, Gwon Y, Song S, Kim SH, Moon SW, Jo DG, Jung YK. FcγRIIb-SHIP2 axis links Aβ to tau pathology by disrupting phosphoinositide metabolism in Alzheimer's disease model. eLife 2016; 5. [PMID: 27834631 PMCID: PMC5106215 DOI: 10.7554/elife.18691] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/17/2016] [Indexed: 02/02/2023] Open
Abstract
Amyloid-β (Aβ)-containing extracellular plaques and hyperphosphorylated tau-loaded intracellular neurofibrillary tangles are neuropathological hallmarks of Alzheimer's disease (AD). Although Aβ exerts neuropathogenic activity through tau, the mechanistic link between Aβ and tau pathology remains unknown. Here, we showed that the FcγRIIb-SHIP2 axis is critical in Aβ1-42-induced tau pathology. Fcgr2b knockout or antagonistic FcγRIIb antibody inhibited Aβ1-42-induced tau hyperphosphorylation and rescued memory impairments in AD mouse models. FcγRIIb phosphorylation at Tyr273 was found in AD brains, in neuronal cells exposed to Aβ1-42, and recruited SHIP2 to form a protein complex. Consequently, treatment with Aβ1-42 increased PtdIns(3,4)P2 levels from PtdIns(3,4,5)P3 to mediate tau hyperphosphorylation. Further, we found that targeting SHIP2 expression by lentiviral siRNA in 3xTg-AD mice or pharmacological inhibition of SHIP2 potently rescued tau hyperphosphorylation and memory impairments. Thus, we concluded that the FcγRIIb-SHIP2 axis links Aβ neurotoxicity to tau pathology by dysregulating PtdIns(3,4)P2 metabolism, providing insight into therapeutic potential against AD.
Collapse
Affiliation(s)
- Tae-In Kam
- Global Research Laboratory, School of Biological Sciences, Seoul National University, Seoul, Korea.,Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Baltimore, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Hyejin Park
- Global Research Laboratory, School of Biological Sciences, Seoul National University, Seoul, Korea.,Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Baltimore, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Youngdae Gwon
- Global Research Laboratory, School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Sungmin Song
- Global Research Laboratory, School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Seo-Hyun Kim
- Global Research Laboratory, School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Seo Won Moon
- Global Research Laboratory, School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Yong-Keun Jung
- Global Research Laboratory, School of Biological Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
186
|
Lee HJ, Ryu JM, Jung YH, Lee SJ, Kim JY, Lee SH, Hwang IK, Seong JK, Han HJ. High glucose upregulates BACE1-mediated Aβ production through ROS-dependent HIF-1α and LXRα/ABCA1-regulated lipid raft reorganization in SK-N-MC cells. Sci Rep 2016; 6:36746. [PMID: 27829662 PMCID: PMC5103190 DOI: 10.1038/srep36746] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/20/2016] [Indexed: 01/07/2023] Open
Abstract
There is an accumulation of evidence indicating that the risk of Alzheimer’s disease is associated with diabetes mellitus, an indicator of high glucose concentrations in blood plasma. This study investigated the effect of high glucose on BACE1 expression and amyloidogenesis in vivo, and we present details of the mechanism associated with those effects. Our results, using ZLC and ZDF rat models, showed that ZDF rats have high levels of amyloid-beta (Aβ), phosphorylated tau, BACE1, and APP-C99. In vitro result with mouse hippocampal neuron and SK-N-MC, high glucose stimulated Aβ secretion and apoptosis in a dose-dependent manner. In addition, high glucose increased BACE1 and APP-C99 expressions, which were reversed by a reactive oxygen species (ROS) scavenger. Indeed, high glucose increased intracellular ROS levels and HIF-1α expression, associated with regulation of BACE1 and Liver X Receptor α (LXRα). In addition, high glucose induced ATP-binding cassette transporter A1 (ABCA1) down-regulation, was associated with LXR-induced lipid raft reorganization and BACE1 localization on the lipid raft. Furthermore, silencing of BACE1 expression was shown to regulate Aβ secretion and apoptosis of SK-N-MC. In conclusion, high glucose upregulates BACE1 expression and activity through HIF-1α and LXRα/ABCA1-regulated lipid raft reorganization, leading to Aβ production and apoptosis of SK-N-MC.
Collapse
Affiliation(s)
- Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Jung Min Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Sei-Jung Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Jeong Yeon Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea.,Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, 330-930, Republic of Korea
| | - In Koo Hwang
- BK21 PLUS Program for Creative Veterinary Science Research, and Research Institute for Veterinary Science; Seoul National University and Korea Mouse Phenotyping Center (KMPC), Seoul, Korea.,Department of Anatomy and Cell Biology; Korea Mouse Phenotyping Center (KMPC); College of Veterinary Medicine; Seoul National University, Seoul, Korea
| | - Je Kyung Seong
- BK21 PLUS Program for Creative Veterinary Science Research, and Research Institute for Veterinary Science; Seoul National University and Korea Mouse Phenotyping Center (KMPC), Seoul, Korea.,Department of Anatomy and Cell Biology; Korea Mouse Phenotyping Center (KMPC); College of Veterinary Medicine; Seoul National University, Seoul, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
187
|
Functional modulation of strychnine-sensitive glycine receptors in rat hippocampal pyramidal neurons by amyloid-β protein (1-42). Brain Res 2016; 1651:61-72. [DOI: 10.1016/j.brainres.2016.09.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/07/2016] [Accepted: 09/09/2016] [Indexed: 11/17/2022]
|
188
|
Shi S, Liang D, Bao M, Xie Y, Xu W, Wang L, Wang Z, Qiao Z. Gx-50 Inhibits Neuroinflammation via α7 nAChR Activation of the JAK2/STAT3 and PI3K/AKT Pathways. J Alzheimers Dis 2016; 50:859-71. [PMID: 26836188 DOI: 10.3233/jad-150963] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recent studies have revealed that the α7 nicotinic acetylcholine receptor (α7 nAChR) is a critical link between inflammation and neurodegeneration, which is closely associated with Alzheimer's disease (AD). The JAK2/STAT3 and PI3K/AKT signaling pathways contribute to the neuroprotective and anti-inflammatory effects of α7nAChR. Our previous studies have shown that treatment with gx-50 improves cognitive function and is neuroprotective. Here, we investigated the effect of gx-50 on α7 nAChR and Aβ-induced inflammation in microglia. First, the binding affinity of gx-50 to α7 nAChR was examined using the fluorescence-based Octet RED system, and the expression of α7 nAChR was detected using real-time PCR and western blotting. We also investigated downstream events of α7 nAChR activity, including the translocation of p-STAT3 and the phosphorylation of JAK2, STAT3, PI3K, and AKT. Finally, the effect of gx-50 on Aβ-induced inflammation via α7 nAChR-mediated signaling pathways was investigated using cytokine assays. The results showed that gx-50 is able to act as a specific ligand to activate α7 nAChR, which then upregulates the JAK2/STAT3 and PI3K/AKT signaling pathways to inhibit the secretions of pro-inflammatory cytokines, such as IL-1β. In conclusion, the results suggest that gx-50 could inhibit the Aβ-induced inflammatory response in microglia via α7 nAChR activity, which might be a successful therapeutic target against AD.
Collapse
|
189
|
Lombardo S, Catteau J, Besson M, Maskos U. A role for β2* nicotinic receptors in a model of local amyloid pathology induced in dentate gyrus. Neurobiol Aging 2016; 46:221-34. [DOI: 10.1016/j.neurobiolaging.2016.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 05/22/2016] [Accepted: 06/09/2016] [Indexed: 12/22/2022]
|
190
|
Lepeta K, Lourenco MV, Schweitzer BC, Martino Adami PV, Banerjee P, Catuara-Solarz S, de La Fuente Revenga M, Guillem AM, Haidar M, Ijomone OM, Nadorp B, Qi L, Perera ND, Refsgaard LK, Reid KM, Sabbar M, Sahoo A, Schaefer N, Sheean RK, Suska A, Verma R, Vicidomini C, Wright D, Zhang XD, Seidenbecher C. Synaptopathies: synaptic dysfunction in neurological disorders - A review from students to students. J Neurochem 2016; 138:785-805. [PMID: 27333343 PMCID: PMC5095804 DOI: 10.1111/jnc.13713] [Citation(s) in RCA: 245] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/03/2016] [Accepted: 06/06/2016] [Indexed: 12/12/2022]
Abstract
Synapses are essential components of neurons and allow information to travel coordinately throughout the nervous system to adjust behavior to environmental stimuli and to control body functions, memories, and emotions. Thus, optimal synaptic communication is required for proper brain physiology, and slight perturbations of synapse function can lead to brain disorders. In fact, increasing evidence has demonstrated the relevance of synapse dysfunction as a major determinant of many neurological diseases. This notion has led to the concept of synaptopathies as brain diseases with synapse defects as shared pathogenic features. In this review, which was initiated at the 13th International Society for Neurochemistry Advanced School, we discuss basic concepts of synapse structure and function, and provide a critical view of how aberrant synapse physiology may contribute to neurodevelopmental disorders (autism, Down syndrome, startle disease, and epilepsy) as well as neurodegenerative disorders (Alzheimer and Parkinson disease). We finally discuss the appropriateness and potential implications of gathering synapse diseases under a single term. Understanding common causes and intrinsic differences in disease-associated synaptic dysfunction could offer novel clues toward synapse-based therapeutic intervention for neurological and neuropsychiatric disorders. In this Review, which was initiated at the 13th International Society for Neurochemistry (ISN) Advanced School, we discuss basic concepts of synapse structure and function, and provide a critical view of how aberrant synapse physiology may contribute to neurodevelopmental (autism, Down syndrome, startle disease, and epilepsy) as well as neurodegenerative disorders (Alzheimer's and Parkinson's diseases), gathered together under the term of synaptopathies. Read the Editorial Highlight for this article on page 783.
Collapse
Affiliation(s)
- Katarzyna Lepeta
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Barbara C Schweitzer
- Department for Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
| | - Pamela V Martino Adami
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir-IIBBA-CONICET, Buenos Aires, Argentina
| | - Priyanjalee Banerjee
- Department of Biochemistry, Institute of Post Graduate Medical Education & Research, Kolkata, West Bengal, India
| | - Silvina Catuara-Solarz
- Systems Biology Program, Cellular and Systems Neurobiology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Mario de La Fuente Revenga
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Alain Marc Guillem
- Laboratorio de Neurotoxicología, Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México D.F. 07000, Mexico
| | - Mouna Haidar
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Omamuyovwi M Ijomone
- Department of Human Anatomy, Cross River University of Technology, Okuku Campus, Cross River, Nigeria
| | - Bettina Nadorp
- The Department of Biological Chemistry, The Edmond and Lily Safra Center for Brain Sciences, The Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, Israel
| | - Lin Qi
- Laboratory of Molecular Neuro-Oncology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, United States of America
| | - Nirma D Perera
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Louise K Refsgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Kimberley M Reid
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Mariam Sabbar
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Arghyadip Sahoo
- Department of Biochemistry, Midnapore Medical College, West Bengal University of Health Sciences, West Bengal, India
| | - Natascha Schaefer
- Institute for Clinical Neurobiology, Julius-Maximilians-University of Wuerzburg, Wuerzburg, Germany
| | - Rebecca K Sheean
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Anna Suska
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Rajkumar Verma
- Department of Neurosciences Uconn Health Center, Farmington, CT, United States of America
| | | | - Dean Wright
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Xing-Ding Zhang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Constanze Seidenbecher
- Department for Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany. .,Center for Behavioral Brain Sciences (CBBS) Magdeburg, Magdeburg, Germany.
| |
Collapse
|
191
|
Hippocampal network dynamics in response to α7 nACh receptors activation in amyloid-β overproducing transgenic mice. Neurobiol Aging 2016; 45:161-168. [DOI: 10.1016/j.neurobiolaging.2016.05.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/26/2016] [Accepted: 05/29/2016] [Indexed: 12/18/2022]
|
192
|
Astroglial calcium signalling in Alzheimer's disease. Biochem Biophys Res Commun 2016; 483:1005-1012. [PMID: 27545605 DOI: 10.1016/j.bbrc.2016.08.088] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/15/2016] [Indexed: 12/14/2022]
Abstract
Neuroglial contribution to Alzheimer's disease (AD) is pathologically relevant and highly heterogeneous. Reactive astrogliosis and activation of microglia contribute to neuroinflammation, whereas astroglial and oligodendroglial atrophy affect synaptic transmission and underlie the overall disruption of the central nervous system (CNS) connectome. Astroglial function is tightly integrated with the intracellular ionic signalling mediated by complex dynamics of cytosolic concentrations of free Ca2+ and Na+. Astroglial ionic signalling is mediated by plasmalemmal ion channels, mainly associated with ionotropic receptors, pumps and solute carrier transporters, and by intracellular organelles comprised of the endoplasmic reticulum and mitochondria. The relative contribution of these molecular cascades/organelles can be plastically remodelled in development and under environmental stress. In AD astroglial Ca2+ signalling undergoes substantial reorganisation due to an abnormal regulation of expression of Ca2+ handling molecular cascades.
Collapse
|
193
|
Hong HS, Maezawa I, Petrlova J, Zhao XY, C Voss J, Jin LW. Tomoregulin (TMEFF2) Binds Alzheimer's Disease Amyloid-β (Aβ) Oligomer and AβPP and Protects Neurons from Aβ-Induced Toxicity. J Alzheimers Dis 2016; 48:731-43. [PMID: 26402097 DOI: 10.3233/jad-150318] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amyloid-β (Aβ) protein causes neurotoxicity and its abnormal aggregation into amyloid is a pathological hallmark of Alzheimer's disease (AD). Cellular proteins able to interact with Aβ or its precursor, AβPP (amyloid-β protein precursor), may regulate Aβ production and neurotoxicity. We identified a brain-enriched type I transmembrane protein, tomoregulin (TR), that directly binds Aβ and Aβ oligomers (AβO). TR co-immunoprecipitated with Aβ and AβO in cultured cells and co-localized with amyloid plaques and intraneuronal Aβ in the 5xFAD AD mouse model. TR was also enriched in astrocytic processes reactive to amyloid plaques. Surface plasmon resonance spectroscopy studies showed that the extracellular domain of TR binds to AβO with a high affinity (KD = 76.8 nM). Electron paramagnetic resonance spectroscopy also demonstrated a physical interaction between spin-labeled Aβ and the TR extracellular domain in solution. Furthermore, TR also interacted with AβPP and enhanced its cleavage by α-secretase. Both cellular expression of TR and application of recombinant TR extracellular domain protected N2a neurons from AβO-induced neuronal death. These data provide first evidence that neuronal and astrocytic expression of TR is intimately related to Aβ metabolism and toxicity, and could be neuroprotective through its direct interaction with Aβ and AβPP.
Collapse
Affiliation(s)
- Hyun-Seok Hong
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, CA, USA
| | - Izumi Maezawa
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, CA, USA.,Alzheimer's Disease Center, University of California Davis Medical Center, Sacramento, CA, USA.,M.I.N.D. (Medical Investigation of Neurodevelopmental Disorders) Institute, University of California Davis Medical Center, Sacramento, CA, USA
| | - Jitka Petrlova
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - Xiao-Yan Zhao
- Biologics Research-USIC, Bayer Healthcare Pharmaceuticals, San Francisco, CA, USA
| | - John C Voss
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, CA, USA.,Alzheimer's Disease Center, University of California Davis Medical Center, Sacramento, CA, USA.,M.I.N.D. (Medical Investigation of Neurodevelopmental Disorders) Institute, University of California Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|
194
|
Agostini M, Fasolato C. When, where and how? Focus on neuronal calcium dysfunctions in Alzheimer's Disease. Cell Calcium 2016; 60:289-298. [PMID: 27451385 DOI: 10.1016/j.ceca.2016.06.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD), since its characterization as a precise form of dementia with its own pathological hallmarks, has captured scientists' attention because of its complexity. The last 30 years have been filled with discoveries regarding the elusive aetiology of this disease and, thanks to advances in molecular biology and live imaging techniques, we now know that an important role is played by calcium (Ca2+). Ca2+, as ubiquitous second messenger, regulates a vast variety of cellular processes, from neuronal excitation and communication, to muscle fibre contraction and hormone secretion, with its action spanning a temporal scale that goes from microseconds to hours. It is therefore very challenging to conceive a single hypothesis that can integrate the numerous findings on this issue with those coming from the classical fields of AD research such as amyloid-beta (Aβ) and tau pathology. In this contribution, we will focus our attention on the Ca2+ hypothesis of AD, dissecting it, as much as possible, in its subcellular localization, where the Ca2+ signal meets its specificity. We will also follow the temporal evolution of the Ca2+ hypothesis, providing some of the most updated discoveries. Whenever possible, we will link the findings regarding Ca2+ dysfunction to the other players involved in AD pathogenesis, hoping to provide a crossover body of evidence, useful to amplify the knowledge that will lead towards the discovery of an effective therapy.
Collapse
Affiliation(s)
- Mario Agostini
- Department of Biomedical Sciences, University of Padua, Italy.
| | | |
Collapse
|
195
|
Kallel EA, Vangel C, Elbaum D. Conformational analysis of 2-substituted piperazines. Bioorg Med Chem Lett 2016; 26:3010-3013. [PMID: 27212066 DOI: 10.1016/j.bmcl.2016.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 11/16/2022]
Abstract
The unusual activity differences of carbon linked versus oxygen linked 2-substituted piperazines as α7 nicotinic acetylcholine receptor agonists led to a conformational study of several examples. The conformational preferences of which are absent from the literature. We report the first study and explanation of the conformational preference of 2-substiturted piperazines and show an example of how this preference controls binding in a pharmaceutically relevant case. In all cases the axial conformation for these 1-acyl and 1 aryl 2-substituted piperazines was found to be preferred. For the ether linked compounds, the axial conformation was found to be further stabilized by an intramolecular hydrogen bond. The axial orientation also places the basic and pyridyl nitrogens into a special orientation that closely mimics nicotine. Molecular modeling studies confirm that the R enantiomers of the compounds can bind to the α7 nicotinic acetylcholine receptor with the basic and pyridyl nitrogens colocalized with their counterparts in Epibatidine.
Collapse
Affiliation(s)
- E Adam Kallel
- Retrophin, Inc. 12255 El Camino Real, Suite 250, San Diego, CA 92130, United States
| | - Colin Vangel
- Retrophin, Inc. 12255 El Camino Real, Suite 250, San Diego, CA 92130, United States
| | - Daniel Elbaum
- Retrophin, Inc, 301 Binney Street, Cambridge, MA 02142, United States
| |
Collapse
|
196
|
Alzheimer disease: modeling an Aβ-centered biological network. Mol Psychiatry 2016; 21:861-71. [PMID: 27021818 DOI: 10.1038/mp.2016.38] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/16/2016] [Accepted: 02/18/2016] [Indexed: 01/15/2023]
Abstract
In genetically complex diseases, the search for missing heritability is focusing on rare variants with large effect. Thanks to next generation sequencing technologies, genome-wide characterization of these variants is now feasible in every individual. However, a lesson from current studies is that collapsing rare variants at the gene level is often insufficient to obtain a statistically significant signal in case-control studies, and that network-based analyses are an attractive complement to classical approaches. In Alzheimer disease (AD), according to the prevalent amyloid cascade hypothesis, the pathology is driven by the amyloid beta (Aβ) peptide. In past years, based on experimental studies, several hundreds of proteins have been shown to interfere with Aβ production, clearance, aggregation or toxicity. Thanks to a manual curation of the literature, we identified 335 genes/proteins involved in this biological network and classified them according to their cellular function. The complete list of genes, or its subcomponents, will be of interest in ongoing AD genetic studies.
Collapse
|
197
|
Weng PH, Chen JH, Chen TF, Sun Y, Wen LL, Yip PK, Chu YM, Chen YC. CHRNA7 Polymorphisms and Dementia Risk: Interactions with Apolipoprotein ε4 and Cigarette Smoking. Sci Rep 2016; 6:27231. [PMID: 27249957 PMCID: PMC4890170 DOI: 10.1038/srep27231] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/15/2016] [Indexed: 12/16/2022] Open
Abstract
α7 nicotinic acetylcholine receptor (α7nAChR, encoded by CHRNA7) is involved in dementia pathogenesis through cholinergic neurotransmission, neuroprotection and interactions with amyloid-β. Smoking promotes atherosclerosis and increases dementia risk, but nicotine exerts neuroprotective effect via α7nAChR in preclinical studies. No studies explored the gene-gene, gene-environment interactions between CHRNA7 polymorphism, apolipoprotein E (APOE) ε4 status and smoking on dementia risk. This case-control study recruited 254 late-onset Alzheimer’s disease (LOAD) and 115 vascular dementia (VaD) cases (age ≥65) from the neurology clinics of three teaching hospitals in Taiwan during 2007–2010. Controls (N = 435) were recruited from health checkup programs and volunteers during the same period. Nine CHRNA7 haplotype-tagging single nucleotide polymorphisms representative for Taiwanese were genotyped. Among APOE ε4 non-carriers, CHRNA7 rs7179008 variant carriers had significantly decreased LOAD risk after correction for multiple tests (GG + AG vs. AA: adjusted odds ratio = 0.29, 95% confidence interval = 0.13–0.64, P = 0.002). Similar findings were observed for carriers of GT haplotype in CHRNA7 block4. A significant interaction was found between rs7179008, GT haplotype in block4 and APOE ε4 on LOAD risk. rs7179008 variant also reduced the detrimental effect of smoking on LOAD risk. No significant association was found between CHRNA7 and VaD. These findings help to understand dementia pathogenesis.
Collapse
Affiliation(s)
- Pei-Hsuan Weng
- Department of Family Medicine, Taiwan Adventist Hospital, Taipei, Taiwan.,Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Jen-Hau Chen
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Geriatrics and Gerontology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu Sun
- Department of Neurology, En Chu Kong Hospital, New Taipei City, Taiwan
| | - Li-Li Wen
- Department of Laboratory Medicine, En Chu Kong Hospital, New Taipei City, Taiwan
| | - Ping-Keung Yip
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan.,Center of Neurological Medicine, Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Yi-Min Chu
- Department of Laboratory Medicine, Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Yen-Ching Chen
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan.,Research Center for Genes, Environment and Human Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
198
|
Chalon S, Vercouillie J, Guilloteau D, Suzenet F, Routier S. PET tracers for imaging brain α7 nicotinic receptors: an update. Chem Commun (Camb) 2016; 51:14826-31. [PMID: 26359819 DOI: 10.1039/c5cc04536c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Positron emission tomography (PET) molecular imaging of brain targets is a powerful tool to diagnose, follow up, and develop treatments and personalized medicine for a number of acute and chronic brain disorders. The availability of β+ emitter tracers labelled with [(11)C] or [(18)F] having optimal characteristics of affinity and selectivity for alpha-7 nicotinic receptors (α7R) has received considerable attention, due to the major implication of these receptors in brain functions. The aim of this review is to identify the interest and need for the in vivo exploration of α7R by PET molecular imaging, which tools are currently available for this and how to progress.
Collapse
Affiliation(s)
- S Chalon
- UMR Inserm U930, Université François-Rabelais de Tours, F-37000 Tours, France.
| | | | | | | | | |
Collapse
|
199
|
Sonkar V, Kulkarni PP, Chaurasia SN, Dash A, Jauhari A, Parmar D, Yadav S, Dash D. Plasma Fibrinogen Is a Natural Deterrent to Amyloid Beta-Induced Platelet Activation. Mol Med 2016; 22:224-232. [PMID: 27262026 DOI: 10.2119/molmed.2016.00003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/02/2016] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder, characterized by extensive loss of neurons, and deposition of amyloid beta (Aβ) in the form of extracellular plaques. Aβ is considered to have critical role in synaptic loss and neuronal death underlying cognitive decline. Platelets contribute to 95% of circulating amyloid-precursor protein that releases Aβ into circulation. We have recently demonstrated that, Aβ active fragment containing amino acid sequence 25-35 (Aβ25-35) is highly thrombogenic in nature, and elicits strong aggregation of washed human platelets in RhoA-dependent manner. In the present study we evaluated the influence of fibrinogen on Aβ-induced platelet activation. Intriguingly, Aβ failed to induce aggregation of platelets suspended in plasma but not in buffer. Fibrinogen brought about dose-dependent decline in aggregatory response of washed human platelets elicited by Aβ25-35, which could be reversed by increasing doses of Aβ. Fibrinogen also attenuated Aβ-induced platelet responses like secretion, clot retraction, rise in cytosolic Ca+2 and reactive oxygen species (ROS). Fibrinogen prevented intracellular accumulation of full length amyloid beta peptide (Aβ42) in platelets as well as neuronal cells. We conclude that fibrinogen serves as a physiological check against the adverse effects of Aβ by preventing its interaction with cells.
Collapse
Affiliation(s)
- Vijay Sonkar
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Paresh P Kulkarni
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Susheel N Chaurasia
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ayusman Dash
- Indian Institute of Science Education and Research, Kolkata, India
| | - Abhishek Jauhari
- Developmental Toxicology Division, Indian Institute of Toxicological Research, Lucknow, Uttar Pradesh, India
| | - Devendra Parmar
- Developmental Toxicology Division, Indian Institute of Toxicological Research, Lucknow, Uttar Pradesh, India
| | - Sanjay Yadav
- Developmental Toxicology Division, Indian Institute of Toxicological Research, Lucknow, Uttar Pradesh, India
| | - Debabrata Dash
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
200
|
Freund RK, Gibson ES, Potter H, Dell'Acqua ML. Inhibition of the Motor Protein Eg5/Kinesin-5 in Amyloid β-Mediated Impairment of Hippocampal Long-Term Potentiation and Dendritic Spine Loss. Mol Pharmacol 2016; 89:552-9. [PMID: 26957206 PMCID: PMC4851299 DOI: 10.1124/mol.115.103085] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/07/2016] [Indexed: 01/09/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by neurofibrillary tangles, amyloid plaques, and neurodegeneration. However, this pathology is preceded by increased soluble amyloid beta (Aβ) 1-42 oligomers that interfere with the glutamatergic synaptic plasticity required for learning and memory, includingN-methyl-d-aspartate receptor (NMDAR)-dependent long-term potentiation (LTP). In particular, soluble Aβ(1-42) acutely inhibits LTP and chronically causes synapse loss. Many mechanisms have been proposed for Aβ-induced synaptic dysfunction, but we recently found that Aβ(1-42) inhibits the microtubule motor protein Eg5/kinesin-5. Here we compared the impacts of Aβ(1-42) and monastrol, a small-molecule Eg5 inhibitor, on LTP in hippocampal slices and synapse loss in neuronal cultures. Acute (20-minute) treatment with monastrol, like Aβ, completely inhibited LTP at doses >100 nM. In addition, 1 nM Aβ(1-42) or 50 nM monastrol inhibited LTP #x223c;50%, and when applied together caused complete LTP inhibition. At concentrations that impaired LTP, neither Aβ(1-42) nor monastrol inhibited NMDAR synaptic responses until #x223c;60 minutes, when only #x223c;25% inhibition was seen for monastrol, indicating that NMDAR inhibition was not responsible for LTP inhibition by either agent when applied for only 20 minutes. Finally, 48 hours of treatment with either 0.5-1.0μM Aβ(1-42) or 1-5μM monastrol reduced the dendritic spine/synapse density in hippocampal cultures up to a maximum of #x223c;40%, and when applied together at maximal concentrations, no additional spine loss resulted. Thus, monastrol can mimic and in some cases occlude the impact of Aβon LTP and synapse loss, suggesting that Aβinduces acute and chronic synaptic dysfunction in part through inhibiting Eg5.
Collapse
Affiliation(s)
- Ronald K Freund
- Department of Pharmacology (R.K.F., E.S.G., M.L.D.'A.), and Department Neurology (H.P.), School of Medicine, and Linda Crnic Institute for Down Syndrome (M.L.D.'A., H.P.), Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - Emily S Gibson
- Department of Pharmacology (R.K.F., E.S.G., M.L.D.'A.), and Department Neurology (H.P.), School of Medicine, and Linda Crnic Institute for Down Syndrome (M.L.D.'A., H.P.), Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - Huntington Potter
- Department of Pharmacology (R.K.F., E.S.G., M.L.D.'A.), and Department Neurology (H.P.), School of Medicine, and Linda Crnic Institute for Down Syndrome (M.L.D.'A., H.P.), Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| | - Mark L Dell'Acqua
- Department of Pharmacology (R.K.F., E.S.G., M.L.D.'A.), and Department Neurology (H.P.), School of Medicine, and Linda Crnic Institute for Down Syndrome (M.L.D.'A., H.P.), Anschutz Medical Campus, University of Colorado, Aurora, Colorado
| |
Collapse
|