Skip to main content
Erschienen in: Neurology and Therapy 1/2017

Open Access 01.07.2017 | Review

The Study of Postmortem Human Synaptosomes for Understanding Alzheimer’s Disease and Other Neurological Disorders: A Review

verfasst von: Jia-Fong Jhou, Hwan-Ching Tai

Erschienen in: Neurology and Therapy | Sonderheft 1/2017

Abstract

Synaptic dysfunction is thought to play important roles in the pathophysiology of many neurological diseases, including Alzheimer’s disease, Parkinson’s disease, and schizophrenia. Over the past few decades, there have been systematic efforts to collect postmortem brain tissues via autopsies, leading to the establishment of dozens of human brain banks around the world. From cryopreserved human brain tissues, it is possible to isolate detached-and-resealed synaptic terminals termed synaptosomes, which remain metabolically and enzymatically active. Synaptosomes have become important model systems for studying human synaptic functions, being much more accessible than ex vivo brain slices or primary neuronal cultures. Here we review recent advances in the establishment of human brain banks, the isolation of synaptosomes, their biological activities, and various analytical techniques for investigating their biochemical and ultrastructural properties. There are unique insights to be gained by directly examining human synaptosomes, which cannot be substituted by animal models. We will also discuss how human synaptosome research has contributed to better understanding of neurological disorders, especially Alzheimer’s disease.
Hinweise

Enhanced content

To view enhanced content for this article go to http://​www.​medengine.​com/​Redeem/​CAD8F06039796B6E​.

Introduction

Within the context of neural networks, the synapse is the region around the point of contact between two neurons, and there are two basic types depending on signal transmission mechanisms: chemical synapses and electrical synapses [1]. There are hundreds of neuron types and possibly thousands of synapse types in the mammalian brain. The number of chemical synapses in the neocortex alone exceeds 100 trillion [2]. It is, therefore, a monumental task to understand the sheer complexity and diversity of synaptic networks in the brain.
A very important method for studying synaptic terminals in the brain is to isolate them from tissues by subcellular fractionation techniques, and these preparations enriched in nerve endings are called “synaptosomes”. Synaptosomes were first isolated in the late 1950s [3], and extensively studied by electron microscopy to understand their structural properties and internal contents [46]. When brain tissues are homogenized, the shear force causes the nerve terminals to be torn apart from the axons and dendrites, and they reseal into spherical structures which can be isolated by centrifugation methods. The isolation procedure is schematically presented in Fig. 1, using excitatory synapses of pyramidal neurons as an example. The presynaptic terminal of the glutamatergic synapse is usually a varicosity (bouton) along the axon, while the postsynaptic terminal is usually a spine protruding from the dendrite. During the homogenization process, the synaptic terminals break off and reseal into vesicular structures around 0.5–1 μm in diameter [7]. By electron microscopy, one may find vesicular objects of synaptic origin, containing synaptic vesicles or postsynaptic density (PSD), as well as membrane-enclosed fragments of myelin, mitochondria, microscomes, lysosomes, and plasma membranes [810].
Synaptosomes freshly prepared from brain tissues are metabolically and enzymatically active. They contain the molecular machinery necessary for the uptake, storage, and release of neurotransmitters, as well as the channels and receptors required for signal transduction. They are active in protein synthesis, maintenance of plasma membrane potential, and ion homeostasis [11]. Synaptosomes can be further fractionated to collect various subcomponents such as synaptic vesicles, postsynaptic densities (PSD), and synaptic plasma membranes [4, 12]. The study of synaptosomes has been one of the cornerstones of neurochemistry research, being instrumental to our understanding of the molecular machinery of neurotransmission and synaptic protein–protein interaction networks [11]. The range of biochemical responses that remain active in synaptosomes continue to expand as more assays are being developed. For instance, recent studies have demonstrated the ability to measure synaptosome insulin response from frozen mouse brains [13], as well as the upregulation and downregulation of SUMO-lylation in mouse synaptosomes by delivering enzymes linked to TAT peptides [14]. While earlier works in synaptosome research used animal brains, a growing number of studies have been directly applied to human brain tissues. In this review we will discuss how to access human brain tissues, how to prepare human synaptosomes, how to study synaptosomes, and what may be learned from such studies. This article is based on previously conducted studies and does not involve any new studies of human or animal subjects performed by any of the authors.

Human Brain Banks

The collection of human brains to study psychiatric and neurological disorders can be traced back to the late nineteenth century, but more systematic approaches toward establishing human brain banks started in the 1960s in the United States [15]. Today, there are dozens of human brain banks around the world dedicated to the collection of postmortem brain tissues after autopsies. Regional consortiums of brain banks, such as BrainNet Europe [16] and the NeuroBioBank [17] under the National Institutes of Health, USA, have been established in order to standardize the diagnosis and tissue collection procedures to provide researchers better access to large cohorts of human subjects.
There are two fundamental questions about the study of synaptosomes using human brain tissues. The first is what can be gained by studying postmortem human tissues compared animal models. The second is the validity of such studies considering the issue of post-mortem interval (PMI) between the time of death and the collection of tissues by autopsy, which is inevitably much longer for human subjects than experimental animals.
The most compelling reason for studying human brain tissues in great detail is to understand the underlying neuropathology of various developmental, traumatic, and degenerative brain disorders. Neuropathology is the basis of accurate diagnosis, etiological understanding, mechanistic insight, and therapeutic development. For example, the identification of β-amyloid fragment in plaques was first made in postmortem frozen tissues [18], as well as the identification of α-synuclein in the Lewy bodies of Parkinson’s disease (PD) [19], tau in frontotemporal dementia (FTD) [20], and TDP-43 in amyotrophic lateral sclerosis (ALS) [21, 22]. A thorough understanding of human neuropathology provides the guiding principles for establishing animal models for various disorders.
It has been shown that, even under healthy conditions, the subcellular localization of huntingtin [23] and α-synuclein [24] are somewhat different in rodent and human brains. This really brings out the question whether current animal models of neurological disorders are truly satisfactory representations of the actual human disease. In the case of Alzheimer’s disease (AD), dozens of animal models have been developed [25, 26], and based on these models tens of billions of US dollars have been invested into finding disease-modifying therapies, but all such efforts have failed over the past decades. We need to address critically to what extent these animal models actually mimic the pathogenesis and pathology of AD in humans, beyond the superficial resemblance of accumulating plaques and tangles, which were artificially driven by overexpressing mutant proteins. For instance, when we first discovered the presence of tau oligomers inside human synaptic terminals [9, 27], we also checked a common animal model of tauopathy, rTg4510 mice overexpressing P301L human mutant tau [28], but its synaptosomes were free of tau oligomers (unpublished data). It is plausible that the mutant tau misfolded so aggressively that they became aggregated before reaching the synapse, causing a marked distinction between mouse and human neuropathology.
There are two basic methods to preserve postmortem brain tissues—formalin fixation and cryopreservation. The former is chemically crosslinked and hence more stable, providing easier handling for histochemical studies. The latter is more amenable to the isolation of various biomolecules and organelles, without the interference of extrinsic chemical modification. To isolate human synaptosomes, cryopreserved tissues are always used. The critical factor affecting the quality of human brain tissues is the PMI [29, 30]. Current trends in brain banking aim to reduce PMI to just a few hours, but for some cases it may still be as long as 24–30 h. Studies have shown that overall rRNA and mRNA stability is maintained for up to 30 h postmortem, without apparent correlation with pH changes due to tissue acidification [31], although specific mRNAs may be selectively degraded under the influence of long PMI or low pH [32]. On the other hand, dephosphorylation may occur on some proteins in less than 1 min, which is a significant problem even in animal experiments [33]. Ideally, the effect of PMI should be individually addressed for each assay condition, but this may not be practical in many experiments.

Isolation of Human Synaptosomes

The isolation of synaptosomes from mammalian brains has evolved relatively little over the last 30 years. This is based on subcellular fractionation techniques similar to those used for isolating other organelles such as mitochondria. In general, tissues are homogenized in a glass-Teflon (Potter–Elvehjem) grinder and an isotonic sucrose buffer (0.32 M). This is followed by low-speed centrifugation (500–1000×g) to remove nuclei and debris, and high-speed centrifugation (10–20K×g) to collect the crude synaptosome pellet (P2 fraction). The P2 fraction may be directly analyzed or further subjected to discontinuous density gradient centrifugation (0.3/0.8/1.2 M sucrose) to remove major contaminants such as myelin fragments and mitochondria, by collecting synaptosomes at the 0.8/1.2 M interface (Fig. 1) [6]. It is possible to collect synaptosomes by either sedimentation or flotation schemes in gradient centrifugation [34, 35]. Even after sucrose gradient centrifugation, the synaptosome faction still contains various organelle contaminants. Many of the vesicular objects from synaptosome preparations do not show clear organelle origin under electron microscopy and it is, therefore, rather difficult to assess quantitatively how presynaptic and postsynaptic terminals are resealed and collected by various isolation schemes.
Based on our experience of examining synaptically enriched preparations with electron microscopy, immunofluorescence microscopy [9, 27], flow cytometry, and super-resolution microscopy (unpublished data), with both human and mouse samples, it appears that at least five classes of nerve ending particles can be found (Fig. 2): (I) intact bipartite synapses, which show snowman-like structures; (II) presynaptic terminals with membrane-free PSD attached; (III) presynaptic terminals with membrane-enclosed PSD, but missing much of the postsynaptic cytoplasm; (IV) isolated presynaptic terminals; (V) isolated postsynaptic terminals. The underlying reason for the formation of class II and III objects is the numerous adhesion molecules linking the active zone to the PSD across the synaptic cleft. One may also occasionally find a single postsynaptic terminals with multiple presynaptic terminals attached, or vice versa.
The relative abundance of different classes of nerve ending particles are presumably affected by homogenization conditions, but detailed quantitative assessments have been very challenging using electron microscopy, immunofluorescence microscopy, and super-resolution techniques. It is generally assumed that isotonic homogenization buffers containing 0.32 M sucrose favor the formation of class II and IV objects. These are conventionally called “synaptosome” preparations, which are used by most laboratories working with isolated synaptic terminals [11]. A less common approach is to prepare isotonic homogenization buffers without sucrose, but with NaCl and other salts. Such preparations are generally called “synaptoneurosomes,” initially described as containing more class I, III, and V objects [36, 37]. In the early years of neurochemistry research, synaptosomes were supposedly presynaptic objects (class II and IV) and neurosomes were supposedly postsynaptic objects (class V), and synaptoneurosomes were supposedly bipartite (class I and III). However, later research revealed that the enrichment of various presynaptic and PSD markers examined by Western blot were hardly distinguishable between synaptosomes and synaptoneurosomes [38]. Our own experiments also confirmed that presynaptic markers (synaptophysin) and postsynaptic markers (PSD-95, MAP2) were similarly enriched in crude synaptosomes vs. synaptoneurosomes (unpublished data). Although sucrose and salt-based buffers may result in somewhat different resealing behavior of the postsynaptic terminal, the difference may not be as striking as initially assumed. Whittaker has suggested that all nerve ending particles obtained by subcellular fractionation may be generally described as “synaptosomes” [11]. On the other hand, “synaptoneurosomes” may be used under a more restricted context to describe synaptic fractions prepared in sucrose-free/high-salt homogenization buffers, which may be more suitable for studying postsynaptic components.
The major drawback of the sucrose gradient method is the change in osmotic pressure. As synaptosomes enter the hypertonic sucrose gradient, they will shrink in size and lose some cytoplasmic components. This can be overcome by using iso-osmotic medium such as Percoll (colloidal silica) [39, 40]. The advantages of Percoll include low viscosity, shorter centrifugation time, and maintaining constant osmolarity to prevent shrinkage/expansion [41]. However, synaptosomes collected from Percoll gradients (at 10/20% interface) are less enriched in synaptic terminals than those collected with sucrose gradients [42]. There is a tradeoff between synaptosome integrity and purity. Another method to carry out synaptosome isolation under isotonic condition is to employ Ficoll dissolved in 0.32 M sucrose [43, 44]. Ficoll is a high-mass, hydrophilic carbohydrate polymer crosslinked into spheres of 2–7 nm in diameter. However, the purity of synaptosomes prepared with Ficoll is also lower than those from sucrose gradients [45]. As such, the optimal choice among sucrose, Percoll, and Ficoll methods may be application-specific.
In our experience, synaptosome preparation protocols developed for rodent brains are equally applicable to postmortem human tissues [9, 27]. The only noticeable difference is that, after grinding brain tissues, human samples contain more insoluble debris than rodent samples, which can be removed by additional centrifugation or 20 μm nylon filters.

Studies of Human Synaptosomes

Studies of postmortem human synaptosomes started in the late 1970s [46]. Around this period, there were numerous studies on synaptosomes freshly prepared from experimental animals to study their morphology, metabolism, and functional activity [11]. Therefore, scientists began to examine the possibility of preparing synaptosomes from postmortem human tissues, comparing them to fresh brain tissues removed during neurosurgeries [4749]. Compared to fresh tissues, synaptosomes prepared from postmortem tissues gradually lost their respiratory activity as PMI increases. Nonetheless, even with a PMI of 24 h, synaptosomes still retained respiratory activity and the ability to release neurotransmitters, and appeared to be morphologically indistinguishable from those from fresh tissues. It was also found that freezing human brain tissues slowly (without additional liquid) and thawing them rapidly were optimal for preserving metabolic activities in synaptosomes [49, 50]. Conversely, rapid freezing, adding sucrose solution as cryo-preservative, and slow thawing were all detrimental, which appeared to be somewhat counterintuitive and the underlying reasons remain unclear.
In the 1970s, researchers began to use postmortem tissues to explore the underlying pathological deficits in neurotransmitter systems in neurodegenerative disorders, especially AD [51]. Because AD is the most common of all neurodegenerative disorders, it was also easier to collect brain tissues from AD patients. In the 1980s, researchers started using synaptosomes prepared from postmortem brains to study AD-associated deficits in neurotransmission, including acetylcholine [52, 53], glutamate [54], and γ-aminobutyric acid (GABA) systems [55]. More recent synaptosome studies have examined the decrease of neprilysin in AD patients [56]. Likewise, the localization of huntingtin has been examined in the synaptosomes of Huntington’s disease patients [23]. For psychiatric disorders, postmortem synaptosome have been applied to study dopamine uptake in schizophrenia patients [57], SNAP-25 levels in bipolar disorder [58], and Na–K-ATPase levels in depressive disorders [59]. Overall, however, there are still very few studies that examine human synaptosomes for neurological disorders other than AD.

Expanding Analytical Techniques

Traditionally, human synaptosome studies have focused on individual aspects of neurotransmission or metabolism. With recent advances in high-throughput techniques such as transcriptomics (by deep RNA sequencing), proteomics (by shotgun mass spectrometry), and metabolomics (by liquid chromatography-tandem mass spectrometry), it should be possible to accelerate the pace of new discoveries with human synaptosomes. Unfortunately, this area of research remains underexplored, with only one preliminary study on the phosphoproteomics of normal human synaptosomes [60]. Moreover, neither transcriptomics nor metabolomics have been applied to study human and animal synaptosomes. The “Omics” approaches may be promising new avenues in human synaptosome research. They may also offer insights for biomarker discovery, because synaptic molecules and proteins are likely to be released into the cerebrospinal fluid or blood, and hence biochemical changes at synapses may be detectable in biological fluids by in vitro diagnostic methods of ever increasing sensitivity. While the presence of many synaptic proteins [61] in the cerebrospinal fluid [62] is well documented, less is known about the release of synaptic proteins into the blood. According to the Plasma Proteome Database [63], many classical synaptic proteins, such as synapotagmin, synaptophysin, SNAP-25, and syntaxin, have been identified in the serum. The serum levels of Aβ-42 and tau are increased in AD patients [64], and both proteins are known to be synaptically released [65, 66], although there may also be non-synaptic routes of their release into the serum.
Another opportunity for high-throughput characterization of synaptosomes is flow cytometry. Gylys and coworkers have shown that immunofluorescence staining in postmortem synaptosomes could be individually isolated or quantified at high speeds [67, 68]. In our experience, tens of thousands of synapses could be quantitatively analyzed in a few minutes on a flow cytometer, and up to three fluorescence channels is feasible for simultaneous detection (unpublished data). Gylys and coworkers have demonstrated that AD patients accumulated Aβ and hyperphosphorylated tau (p-tau) at synaptic sites in multiple brain regions, accompanied by increased synaptosome size, reduced numbers of PSD, and increased free cholesterol [6971]. Co-localization of Aβ and p-tau was demonstrated by dual labeling experiments, and the synaptic accumulation of Aβ occured at the earliest plaque stages, before the accumulation of synaptic p-tau, which appeared at later stages [71, 72]. However, even in early disease stages, p-tau appeared to be elevated in individual Aβ-positive terminals. These results implied that soluble oligomers of Aβ may trigger tau pathology at synaptic terminals, which may be important for the onset of dementia [72]. They have also shown that c-terminal truncated tau was released by presynaptic terminals when postmortem synaptosomes were stimulated by depolarization, supporting the notion that tauopathy may spread across different regions through synaptic networks [66]. Flow cytometry of postmortem synaptosomes have also shown that dopamine transporter was decreased in the putamen region of dementia with Lewy body patients [73].
With the spatial resolution of current flow cytometers, it is not yet possible to differentiate proteins of presynaptic and postsynaptic origins. This differentiation is also very difficult in immunohistochemistry experiments because human tissues have much longer PMI than animals, resulting in less optimal preservation and hence higher background staining. Tai et al. have demonstrated that isolated human synaptic terminals fixed over glass slides could be immunostained to distinguish presynaptic from postsynaptic protein localization [27]. They discovered that normal human tau protein under normal conditions were equally abundant at presynaptic and postsynaptic terminals, against the common misconception that tau was predominantly presynaptic in healthy neurons. Under AD conditions, misfolded and hyperphosphorylated tau proteins were also equally abundant at presynaptic and postsynaptic terminals [9], which lent support to the hypothesis that misfolded tau may transmit trans-synaptically via “prion-like” mechanisms [74, 75]. Figure 3 shows the confocal image of an isolated human synaptosome immunostained with presynaptic and postsynaptic markers. A third immunofluorescence channel can be added to localize the target protein to presynaptic or postsynaptic sites. In fact, immunofluorescence imaging of fixed synaptosomes is technically similar than visualizing synaptic terminals in cultured neurons [9, 27]. Therefore, it is also possible to study synaptosomes by high-content imaging methods incorporating automated microscopes as well as automated image processing [76].
The practical x–y resolution of confocal and epifluorescence microscopy is limited by diffraction to 150–200 nm. Compared to the size of synaptic terminals (500–1000 nm), this resolution is insufficient to reveal protein colocalization within the synapse. The application of direct stochastic optical reconstruction microscopy (dSTORM), a super-resolution microscopy technique based on immunofluorescence, can push the resolution limit down to 30 nm and reveal ultrastructural information within the synapse [77]. The dSTORM technique has yet to be applied to human synaptosomes, but it holds great promise for studying the ultrastructural distribution and colocalization of synaptic proteins.
There are multiple reasons as to why recent studies on human synaptosomes have focused on AD more than any other neurological disorder. First, AD is the most common of all neurodegenerative disorders and occurs frequently in the elderly. Therefore it is easier to build up large cohorts of AD patients in brain banks. Second, AD has often been attributed as a synaptic disorder [78]. Cognitive decline in AD patients shows stronger correlation with synapse loss than with neuronal loss, plaque deposition, tangles accumulation, or various measures of Aβ and tau misfolding [79, 80]. Third, key proteins in AD pathogenesis, including APP, Aβ, ApoE, and tau, have all been observed within human synaptosomes [9, 27, 71, 8183]. Both Aβ and tau are secreted at human synaptic terminals upon activation or stimulation [65, 66]. The transmission of abnormal tau along synaptic networks has been demonstrated in several animal models [8487], and its propagation into cortical regions is accelerated by misfolded Aβ [88]. We have also shown that hyperphosphorylated tau oligomers accumulated at synapses in AD subjects [9, 27], and subsequently observed that, among people with high plaque and tangle pathologies, those who did not accumulate tau oligomers at synaptic sites tended to remian congnitively normal, while those who did generally exhibited the congnitive symptoms of AD  [89]. Therefore, the study of synaptic abnormalities is crucial for understanding the pathogenesis and progression of AD.
Moreover, current therapeutic agents for AD are also focused on preserving synaptic function. There are currently four FDA-approved drugs on the market for AD [90]. Three of them are cholinesterase inhibitors (donepezil, rivastigmine, galantamine) and the other is an N-methyl-d-aspartate receptor antagonist (memantine). The efficacy of cholinesterase inhibitors is explained by the massive degeneration of cholinergic neurons located at the nucleus basalis of Meynert in AD patients [91, 92]. However, currently available drugs only offer symptomatic relief for AD patients, without slowing down the progressive nature of neurodegeneration. Apparently, we need to further investigate the pathophysiology of synapses in AD patients, using the expanding analytical approaches mentioned above, to help us develop disease-modifying therapies which could slow down synaptic and neuronal loss.

Conclusion

The ultimate goal of neuroscience is to understand the human brain. However, due to practical limitations, most of the neuroscience research at the cellular and molecular levels have been conducted with animal models or cultured cells. The growing collection of tissues deposited in human brain banks provides tremendous opportunities for future research to directly investigate the human brain. This line of research will be most valuable for understanding human neurological disorders, especially for studying the “fine pathology” which has been largely overlooked. The “fine pathology” refers to a series of molecular and subcellular changes undetectable by conventional histology techniques, which may very well be quite different in human brains compared to animal models. The only way to tell if they are different is to apply the latest technologies and carefully examine both human and animal brain tissues.
The synapse plays crucial roles in the pathogenesis and pathology of many neurological disorders. The methods of isolating synaptosomes from animal tissues are equally applicable to human tissues. Human synaptosomes can be used to study neurotransmission, metabolism, protein synthesis, protein localization, and posttranslational modification. An increasing number of studies have been examining the synaptosomes of AD patients, and these analyses should be extended to other neurodegenerative and psychiatric disorders. To accelerate the pace of future discoveries, high-throughput analyses including transcriptomics, proteomics, metabolomics, high-content imaging, and flow cytometry should be increasingly applied to human synaptosomes. Ultrastructural studies based on super-resolution fluorescence imaging will offer important avenues for exploration, likely to reveal many exciting discoveries. We are just entering the dawn of human neuroscience and many new discoveries will undoubtedly originate from the direct examination of the human brain.

Acknowledgements

This work was supported by Grant 105-2113-M-002-016 from Ministry of Science and Technology, Taiwan. No funding or sponsorship was received for the publication of this article. All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this manuscript, take responsibility for the integrity of the work as a whole, and have given final approval for the version to be published.

Disclosures

Jia-Fong Jhou and Hwan-Ching Tai have nothing to disclose.

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not involve any new studies of human or animal subjects performed by any of the authors.

Open Access

This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License (http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (https://​creativecommons.​org/​licenses/​by/​4.​0), which permits use, duplication, adaptation, distribution, and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Literatur
1.
Zurück zum Zitat Eccles JC. The synapse: from electrical to chemical transmission. Annu Rev Neurosci. 1982;5:325–39.CrossRefPubMed Eccles JC. The synapse: from electrical to chemical transmission. Annu Rev Neurosci. 1982;5:325–39.CrossRefPubMed
2.
Zurück zum Zitat Tang Y, Nyengaard JR, De Groot DM, Gundersen HJ. Total regional and global number of synapses in the human brain neocortex. Synapse. 2001;41:258–73.CrossRefPubMed Tang Y, Nyengaard JR, De Groot DM, Gundersen HJ. Total regional and global number of synapses in the human brain neocortex. Synapse. 2001;41:258–73.CrossRefPubMed
4.
Zurück zum Zitat Whittaker V, Michaelson I, Kirkland RJA. The separation of synaptic vesicles from nerve-ending particles (‘synaptosomes’). Biochem J. 1964;90:293–303.CrossRefPubMedPubMedCentral Whittaker V, Michaelson I, Kirkland RJA. The separation of synaptic vesicles from nerve-ending particles (‘synaptosomes’). Biochem J. 1964;90:293–303.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat De Robertis E, De Iraldi AP, Garnaiz GRDL, Salganicoff L. Cholinergic and non-cholinergic nerve endings in rat brain. I. Isolation and subcellular distribution of acetylcholine and acetylcholinesterase. J Neurochem. 1962;9:23–35.CrossRef De Robertis E, De Iraldi AP, Garnaiz GRDL, Salganicoff L. Cholinergic and non-cholinergic nerve endings in rat brain. I. Isolation and subcellular distribution of acetylcholine and acetylcholinesterase. J Neurochem. 1962;9:23–35.CrossRef
6.
Zurück zum Zitat Gray E, Whittaker V. The isolation of nerve endings from brain: an electron microscopic study of cell fragments derived by homogenization and centrifugation. J Anat. 1962;96:79–88.PubMedPubMedCentral Gray E, Whittaker V. The isolation of nerve endings from brain: an electron microscopic study of cell fragments derived by homogenization and centrifugation. J Anat. 1962;96:79–88.PubMedPubMedCentral
7.
Zurück zum Zitat Whittaker V. The morphology of fractions of rat forebrain synaptosomes separated on continuous sucrose density gradients. Biochem J. 1968;106:412.CrossRefPubMedPubMedCentral Whittaker V. The morphology of fractions of rat forebrain synaptosomes separated on continuous sucrose density gradients. Biochem J. 1968;106:412.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Jones DG, Revell E. The postnatal development of the synapse: a morphological approach utilizing synaptosomes. I. General features. Z Zellforsch Mikrosk Anat. 1970;111:179–94.CrossRefPubMed Jones DG, Revell E. The postnatal development of the synapse: a morphological approach utilizing synaptosomes. I. General features. Z Zellforsch Mikrosk Anat. 1970;111:179–94.CrossRefPubMed
9.
Zurück zum Zitat Tai HC, Wang BY, Serrano Pozo A, Frosch MP, Spires-Jones TL, Hyman BT. Frequent and symmetric deposition of misfolded tau oligomers within presynaptic and postsynaptic terminals in Alzheimer inverted question marks disease. Acta Neuropathol Commun. 2014;2:146.PubMedPubMedCentral Tai HC, Wang BY, Serrano Pozo A, Frosch MP, Spires-Jones TL, Hyman BT. Frequent and symmetric deposition of misfolded tau oligomers within presynaptic and postsynaptic terminals in Alzheimer inverted question marks disease. Acta Neuropathol Commun. 2014;2:146.PubMedPubMedCentral
10.
Zurück zum Zitat Cotman C, Brown DH, Harrell BW, Anderson NG. Analytical differential centrifugation: an analysis of the sedimentation properties of synaptosomes, mitochondria and lysosomes from rat brain homogenates. Arch Biochem Biophys. 1970;136:436–47.CrossRefPubMed Cotman C, Brown DH, Harrell BW, Anderson NG. Analytical differential centrifugation: an analysis of the sedimentation properties of synaptosomes, mitochondria and lysosomes from rat brain homogenates. Arch Biochem Biophys. 1970;136:436–47.CrossRefPubMed
11.
12.
Zurück zum Zitat Bermejo MK, Milenkovic M, Salahpour A, Ramsey AJ. Preparation of synaptic plasma membrane and postsynaptic density proteins using a discontinuous sucrose gradient. J Vis Exp. 2014. doi:10.3791/51896.PubMedPubMedCentral Bermejo MK, Milenkovic M, Salahpour A, Ramsey AJ. Preparation of synaptic plasma membrane and postsynaptic density proteins using a discontinuous sucrose gradient. J Vis Exp. 2014. doi:10.​3791/​51896.PubMedPubMedCentral
13.
Zurück zum Zitat Franklin W, Taglialatela G. A method to determine insulin responsiveness in synaptosomes isolated from frozen brain tissue. J Neurosci Methods. 2016;261:128–34.CrossRefPubMedPubMedCentral Franklin W, Taglialatela G. A method to determine insulin responsiveness in synaptosomes isolated from frozen brain tissue. J Neurosci Methods. 2016;261:128–34.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Marcelli S, Ficulle E, Iannuzzi F, Kövari E, Nisticò R, Feligioni M. Targeting SUMO-1ylation contrasts synaptic dysfunction in a mouse model of Alzheimer’s disease. Mol Neurobiol. 2016. doi:10.1007/s12035-016-0176-9.PubMed Marcelli S, Ficulle E, Iannuzzi F, Kövari E, Nisticò R, Feligioni M. Targeting SUMO-1ylation contrasts synaptic dysfunction in a mouse model of Alzheimer’s disease. Mol Neurobiol. 2016. doi:10.​1007/​s12035-016-0176-9.PubMed
15.
Zurück zum Zitat Kretzschmar H. Brain banking: opportunities, challenges and meaning for the future. Nat Rev Neurosci. 2009;10:70–8.CrossRefPubMed Kretzschmar H. Brain banking: opportunities, challenges and meaning for the future. Nat Rev Neurosci. 2009;10:70–8.CrossRefPubMed
16.
Zurück zum Zitat Bell JE, Alafuzoff I, Al-Sarraj S, Arzberger T, Bogdanovic N, Budka H, et al. Management of a twenty-first century brain bank: experience in the BrainNet Europe consortium. Acta Neuropathol. 2008;115:497–507.CrossRefPubMed Bell JE, Alafuzoff I, Al-Sarraj S, Arzberger T, Bogdanovic N, Budka H, et al. Management of a twenty-first century brain bank: experience in the BrainNet Europe consortium. Acta Neuropathol. 2008;115:497–507.CrossRefPubMed
17.
Zurück zum Zitat Nichols L, Freund M, Ng C, Kau A, Parisi M, Taylor A, et al. The National Institutes of Health Neurobiobank: a federated national network of human brain and tissue repositories. Biol Psychiatry. 2014;75:e21–2.CrossRefPubMed Nichols L, Freund M, Ng C, Kau A, Parisi M, Taylor A, et al. The National Institutes of Health Neurobiobank: a federated national network of human brain and tissue repositories. Biol Psychiatry. 2014;75:e21–2.CrossRefPubMed
18.
Zurück zum Zitat Glenner GG, Wong CW. Alzheimer’s disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun. 1984;120:885–90.CrossRefPubMed Glenner GG, Wong CW. Alzheimer’s disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun. 1984;120:885–90.CrossRefPubMed
19.
Zurück zum Zitat Spillantini MG, Schmidt ML, Lee VM-Y, Trojanowski JQ, Jakes R, Goedert M. α-Synuclein in Lewy bodies. Nature. 1997;388:839–40.CrossRefPubMed Spillantini MG, Schmidt ML, Lee VM-Y, Trojanowski JQ, Jakes R, Goedert M. α-Synuclein in Lewy bodies. Nature. 1997;388:839–40.CrossRefPubMed
20.
Zurück zum Zitat Poorkaj P, Bird TD, Wijsman E, Nemens E, Garruto RM, Anderson L, et al. Tau is a candidate gene for chromosome 17 frontotemporal dementia. Ann Neurol. 1998;43:815–25.CrossRefPubMed Poorkaj P, Bird TD, Wijsman E, Nemens E, Garruto RM, Anderson L, et al. Tau is a candidate gene for chromosome 17 frontotemporal dementia. Ann Neurol. 1998;43:815–25.CrossRefPubMed
21.
Zurück zum Zitat Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–3.CrossRefPubMed Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–3.CrossRefPubMed
22.
Zurück zum Zitat Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319:1668–72.CrossRefPubMed Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319:1668–72.CrossRefPubMed
23.
Zurück zum Zitat Wood JD, MacMillan JC, Harper PS, Lowenstein PR, Jones AL. Partial characterisation of murine huntingtin and apparent variations in the subcellular localisation of huntingtin in human, mouse and rat brain. Hum Mol Genet. 1996;5:481–7.CrossRefPubMed Wood JD, MacMillan JC, Harper PS, Lowenstein PR, Jones AL. Partial characterisation of murine huntingtin and apparent variations in the subcellular localisation of huntingtin in human, mouse and rat brain. Hum Mol Genet. 1996;5:481–7.CrossRefPubMed
24.
Zurück zum Zitat Kahle PJ, Neumann M, Ozmen L, Müller V, Jacobsen H, Schindzielorz A, et al. Subcellular localization of wild-type and Parkinson’s disease-associated mutant α-synuclein in human and transgenic mouse brain. J Neurosci. 2000;20:6365–73.PubMed Kahle PJ, Neumann M, Ozmen L, Müller V, Jacobsen H, Schindzielorz A, et al. Subcellular localization of wild-type and Parkinson’s disease-associated mutant α-synuclein in human and transgenic mouse brain. J Neurosci. 2000;20:6365–73.PubMed
25.
Zurück zum Zitat Gotz J, Ittner LM. Animal models of Alzheimer’s disease and frontotemporal dementia. Nat Rev Neurosci. 2008;9:532–44.CrossRefPubMed Gotz J, Ittner LM. Animal models of Alzheimer’s disease and frontotemporal dementia. Nat Rev Neurosci. 2008;9:532–44.CrossRefPubMed
26.
Zurück zum Zitat Cuadrado-Tejedor M, García-Osta A. Current animal models of Alzheimer’s disease: challenges in translational research. Front Neurol. 2014;5:182.CrossRefPubMedPubMedCentral Cuadrado-Tejedor M, García-Osta A. Current animal models of Alzheimer’s disease: challenges in translational research. Front Neurol. 2014;5:182.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Tai HC, Serrano-Pozo A, Hashimoto T, Frosch MP, Spires-Jones TL, Hyman BT. The synaptic accumulation of hyperphosphorylated tau oligomers in Alzheimer disease is associated with dysfunction of the ubiquitin-proteasome system. Am J Pathol. 2012;181:1426–35.CrossRefPubMedPubMedCentral Tai HC, Serrano-Pozo A, Hashimoto T, Frosch MP, Spires-Jones TL, Hyman BT. The synaptic accumulation of hyperphosphorylated tau oligomers in Alzheimer disease is associated with dysfunction of the ubiquitin-proteasome system. Am J Pathol. 2012;181:1426–35.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Santacruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science. 2005;309:476–81.CrossRefPubMedPubMedCentral Santacruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science. 2005;309:476–81.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Blair JA, Wang C, Hernandez D, Siedlak SL, Rodgers MS, Achar RK, et al. Individual case analysis of postmortem interval time on brain tissue preservation. PLoS One. 2016;11:e0151615.CrossRefPubMedPubMedCentral Blair JA, Wang C, Hernandez D, Siedlak SL, Rodgers MS, Achar RK, et al. Individual case analysis of postmortem interval time on brain tissue preservation. PLoS One. 2016;11:e0151615.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Stan AD, Ghose S, Gao XM, Roberts RC, Lewis-Amezcua K, Hatanpaa KJ, et al. Human postmortem tissue: what quality markers matter? Brain Res. 2006;1123:1–11.CrossRefPubMedPubMedCentral Stan AD, Ghose S, Gao XM, Roberts RC, Lewis-Amezcua K, Hatanpaa KJ, et al. Human postmortem tissue: what quality markers matter? Brain Res. 2006;1123:1–11.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Ervin JF, Heinzen EL, Cronin KD, Goldstein D, Szymanski MH, Burke JR, et al. Postmortem delay has minimal effect on brain RNA integrity. J Neuropathol Exp Neurol. 2007;66:1093–9.CrossRefPubMed Ervin JF, Heinzen EL, Cronin KD, Goldstein D, Szymanski MH, Burke JR, et al. Postmortem delay has minimal effect on brain RNA integrity. J Neuropathol Exp Neurol. 2007;66:1093–9.CrossRefPubMed
32.
Zurück zum Zitat Hynd MR, Lewohl JM, Scott HL, Dodd PR. Biochemical and molecular studies using human autopsy brain tissue. J Neurochem. 2003;85:543–62.CrossRefPubMed Hynd MR, Lewohl JM, Scott HL, Dodd PR. Biochemical and molecular studies using human autopsy brain tissue. J Neurochem. 2003;85:543–62.CrossRefPubMed
33.
Zurück zum Zitat Ahmed MM, Gardiner KJ. Preserving protein profiles in tissue samples: differing outcomes with and without heat stabilization. J Neurosci Methods. 2011;196:99–106.CrossRefPubMedPubMedCentral Ahmed MM, Gardiner KJ. Preserving protein profiles in tissue samples: differing outcomes with and without heat stabilization. J Neurosci Methods. 2011;196:99–106.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Dodd P, Hardy J, Oakley A, Edwardson J, Perry E, Delaunoy J-P. A rapid method for preparing synaptosomes: comparison, with alternative procedures. Brain Res. 1981;226:107–18.CrossRefPubMed Dodd P, Hardy J, Oakley A, Edwardson J, Perry E, Delaunoy J-P. A rapid method for preparing synaptosomes: comparison, with alternative procedures. Brain Res. 1981;226:107–18.CrossRefPubMed
35.
Zurück zum Zitat Lathia D, Wesemann W. Serotonin uptake and release by biochemically characterized nerve endings isolated from rat brain by concomitant flotation and sedimentation centrifugation. J Neural Transm. 1975;37:111–26.CrossRefPubMed Lathia D, Wesemann W. Serotonin uptake and release by biochemically characterized nerve endings isolated from rat brain by concomitant flotation and sedimentation centrifugation. J Neural Transm. 1975;37:111–26.CrossRefPubMed
36.
Zurück zum Zitat Hollingsworth EB, McNeal ET, Burton JL, Williams RJ, Daly JW, Creveling CR. Biochemical characterization of a filtered synaptoneurosome preparation from guinea pig cerebral cortex: cyclic adenosine 3′:5′-monophosphate-generating systems, receptors, and enzymes. J Neurosci. 1985;5:2240–53.PubMed Hollingsworth EB, McNeal ET, Burton JL, Williams RJ, Daly JW, Creveling CR. Biochemical characterization of a filtered synaptoneurosome preparation from guinea pig cerebral cortex: cyclic adenosine 3′:5′-monophosphate-generating systems, receptors, and enzymes. J Neurosci. 1985;5:2240–53.PubMed
37.
Zurück zum Zitat Schwartz RD, Skolnick P, Hollingsworth EB, Paul SM. Barbiturate and picrotoxin-sensitive chloride efflux in rat cerebral cortical synaptoneurosomes. FEBS Lett. 1984;175:193–6.CrossRefPubMed Schwartz RD, Skolnick P, Hollingsworth EB, Paul SM. Barbiturate and picrotoxin-sensitive chloride efflux in rat cerebral cortical synaptoneurosomes. FEBS Lett. 1984;175:193–6.CrossRefPubMed
38.
Zurück zum Zitat Johnson MW, Chotiner JK, Watson JB. Isolation and characterization of synaptoneurosomes from single rat hippocampal slices. J Neurosci Methods. 1997;77:151–6.CrossRefPubMed Johnson MW, Chotiner JK, Watson JB. Isolation and characterization of synaptoneurosomes from single rat hippocampal slices. J Neurosci Methods. 1997;77:151–6.CrossRefPubMed
39.
Zurück zum Zitat Lagercrantz H, Pertoft H. Separation of catecholamine storing synaptosomes in colloidal silica density gradients. J Neurochem. 1972;19:811–23.CrossRefPubMed Lagercrantz H, Pertoft H. Separation of catecholamine storing synaptosomes in colloidal silica density gradients. J Neurochem. 1972;19:811–23.CrossRefPubMed
40.
Zurück zum Zitat Nagy A, Delgado-Escueta AV. Rapid preparation of synaptosomes from mammalian brain using nontoxic isoosmotic gradient material (Percoll). J Neurochem. 1984;43:1114–23.CrossRefPubMed Nagy A, Delgado-Escueta AV. Rapid preparation of synaptosomes from mammalian brain using nontoxic isoosmotic gradient material (Percoll). J Neurochem. 1984;43:1114–23.CrossRefPubMed
41.
Zurück zum Zitat Dunkley PR, Jarvie PE, Heath JW, Kidd GJ, Rostas JA. A rapid method for isolation of synaptosomes on Percoll gradients. Brain Res. 1986;372:115–29.CrossRefPubMed Dunkley PR, Jarvie PE, Heath JW, Kidd GJ, Rostas JA. A rapid method for isolation of synaptosomes on Percoll gradients. Brain Res. 1986;372:115–29.CrossRefPubMed
42.
Zurück zum Zitat Tenreiro P, Rebelo S, Martins F, Santos M, Coelho E, Almeida M, et al. Comparison of simple sucrose and percoll based methodologies for synaptosome enrichment. Anal Biochem. 2017;517:1–8.CrossRefPubMed Tenreiro P, Rebelo S, Martins F, Santos M, Coelho E, Almeida M, et al. Comparison of simple sucrose and percoll based methodologies for synaptosome enrichment. Anal Biochem. 2017;517:1–8.CrossRefPubMed
43.
Zurück zum Zitat Booth RF, Clark JB. A rapid method for the preparation of relatively pure metabolically competent synaptosomes from rat brain. Biochem J. 1978;176:365–70.CrossRefPubMedPubMedCentral Booth RF, Clark JB. A rapid method for the preparation of relatively pure metabolically competent synaptosomes from rat brain. Biochem J. 1978;176:365–70.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Kurokawa M, Kato M, Sakamoto T. Distribution of sodium-plus-potassium-stimulated adenosine-triphosphatase activity in isolated nerve-ending particles. Biochem J. 1965;97:833–44.CrossRefPubMedPubMedCentral Kurokawa M, Kato M, Sakamoto T. Distribution of sodium-plus-potassium-stimulated adenosine-triphosphatase activity in isolated nerve-ending particles. Biochem J. 1965;97:833–44.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Joo F, Karnushina I. Morphometric assessment of the composition of the synaptosomal fractions obtained by the use of Ficoll gradients. J Neurochem. 1975;24:839–40.CrossRefPubMed Joo F, Karnushina I. Morphometric assessment of the composition of the synaptosomal fractions obtained by the use of Ficoll gradients. J Neurochem. 1975;24:839–40.CrossRefPubMed
46.
Zurück zum Zitat Klemm N, Kuhar MJ. Post-mortem changes in high affinity choline uptake. J Neurochem. 1979;32:1487–94.CrossRefPubMed Klemm N, Kuhar MJ. Post-mortem changes in high affinity choline uptake. J Neurochem. 1979;32:1487–94.CrossRefPubMed
47.
Zurück zum Zitat Dodd P, Hardy JA, Oakley AE, Strong AJ. Synaptosomes prepared from fresh human cerebral cortex; morphology, respiration and release of transmitter amino acids. Brain Res. 1981;224:419–25.CrossRefPubMed Dodd P, Hardy JA, Oakley AE, Strong AJ. Synaptosomes prepared from fresh human cerebral cortex; morphology, respiration and release of transmitter amino acids. Brain Res. 1981;224:419–25.CrossRefPubMed
48.
Zurück zum Zitat Hardy JA, Dodd PR, Oakley AE, Kidd AM, Perry RH, Edwardson JA. Use of post-mortem human synaptosomes for studies of metabolism and transmitter amino acid release. Neurosci Lett. 1982;33:317–22.CrossRefPubMed Hardy JA, Dodd PR, Oakley AE, Kidd AM, Perry RH, Edwardson JA. Use of post-mortem human synaptosomes for studies of metabolism and transmitter amino acid release. Neurosci Lett. 1982;33:317–22.CrossRefPubMed
49.
Zurück zum Zitat Hardy JA, Dodd PR, Oakley AE, Perry RH, Edwardson JA, Kidd AM. Metabolically active synaptosomes can be prepared from frozen rat and human brain. J Neurochem. 1983;40:608–14.CrossRefPubMed Hardy JA, Dodd PR, Oakley AE, Perry RH, Edwardson JA, Kidd AM. Metabolically active synaptosomes can be prepared from frozen rat and human brain. J Neurochem. 1983;40:608–14.CrossRefPubMed
50.
Zurück zum Zitat Dodd P, Hardy J, Baig E, Kidd A, Bird E, Watson W, et al. Optimization of freezing, storage, and thawing conditions for the preparation of metabolically active synaptosomes from frozen rat and human brain. Neurochem Pathol. 1986;4:177–98.CrossRefPubMed Dodd P, Hardy J, Baig E, Kidd A, Bird E, Watson W, et al. Optimization of freezing, storage, and thawing conditions for the preparation of metabolically active synaptosomes from frozen rat and human brain. Neurochem Pathol. 1986;4:177–98.CrossRefPubMed
51.
Zurück zum Zitat Hardy J, Adolfsson R, Alafuzoff I, Bucht G, Marcusson J, Nyberg P, et al. Transmitter deficits in Alzheimer’s disease. Neurochem Int. 1985;7:545–63.CrossRefPubMed Hardy J, Adolfsson R, Alafuzoff I, Bucht G, Marcusson J, Nyberg P, et al. Transmitter deficits in Alzheimer’s disease. Neurochem Int. 1985;7:545–63.CrossRefPubMed
52.
Zurück zum Zitat Nordberg A, Winblad B. Reduced number of [3H] nicotine and [3H] acetylcholine binding sites in the frontal cortex of Alzheimer brains. Neurosci Lett. 1986;72:115–20.CrossRefPubMed Nordberg A, Winblad B. Reduced number of [3H] nicotine and [3H] acetylcholine binding sites in the frontal cortex of Alzheimer brains. Neurosci Lett. 1986;72:115–20.CrossRefPubMed
53.
Zurück zum Zitat Rylett R, Ball M, Colhoun E. Evidence for high affinity choline transport in synaptosomes prepared from hippocampus and neocortex of patients with Alzheimer’s disease. Brain Res. 1983;289:169–75.CrossRefPubMed Rylett R, Ball M, Colhoun E. Evidence for high affinity choline transport in synaptosomes prepared from hippocampus and neocortex of patients with Alzheimer’s disease. Brain Res. 1983;289:169–75.CrossRefPubMed
54.
Zurück zum Zitat Hardy J, Cowburn R, Barton A, Reynolds G, Lofdahl E, O’Carroll A-M, et al. Region-specific loss of glutamate innervation in Alzheimer’s disease. Neurosci Lett. 1987;73:77–80.CrossRefPubMed Hardy J, Cowburn R, Barton A, Reynolds G, Lofdahl E, O’Carroll A-M, et al. Region-specific loss of glutamate innervation in Alzheimer’s disease. Neurosci Lett. 1987;73:77–80.CrossRefPubMed
55.
Zurück zum Zitat Hardy J, Cowburn R, Barton A, Reynolds G, Dodd P, Wester P, et al. A disorder of cortical GABAergic innervation in Alzheimer’s disease. Neurosci Lett. 1987;73:192–6.CrossRefPubMed Hardy J, Cowburn R, Barton A, Reynolds G, Dodd P, Wester P, et al. A disorder of cortical GABAergic innervation in Alzheimer’s disease. Neurosci Lett. 1987;73:192–6.CrossRefPubMed
56.
Zurück zum Zitat Wang D-S, Lipton RB, Katz MJ, Davies P, Buschke H, Kuslansky G, et al. Decreased neprilysin immunoreactivity in Alzheimer disease, but not in pathological aging. J Neuropathol Exp Neurol. 2005;64:378–85.CrossRefPubMed Wang D-S, Lipton RB, Katz MJ, Davies P, Buschke H, Kuslansky G, et al. Decreased neprilysin immunoreactivity in Alzheimer disease, but not in pathological aging. J Neuropathol Exp Neurol. 2005;64:378–85.CrossRefPubMed
57.
Zurück zum Zitat Haberland N, Hetey L. Studies in postmortem dopamine uptake. II. Alterations of the synaptosomal catecholamine uptake in postmortem brain regions in schizophrenia. J Neural Transm. 1987;68:303–13.CrossRefPubMed Haberland N, Hetey L. Studies in postmortem dopamine uptake. II. Alterations of the synaptosomal catecholamine uptake in postmortem brain regions in schizophrenia. J Neural Transm. 1987;68:303–13.CrossRefPubMed
58.
Zurück zum Zitat Scarr E, Gray L, Keriakous D, Robinson P, Dean B. Increased levels of SNAP-25 and synaptophysin in the dorsolateral prefrontal cortex in bipolar I disorder. Bipolar Disord. 2006;8:133–43.CrossRefPubMed Scarr E, Gray L, Keriakous D, Robinson P, Dean B. Increased levels of SNAP-25 and synaptophysin in the dorsolateral prefrontal cortex in bipolar I disorder. Bipolar Disord. 2006;8:133–43.CrossRefPubMed
59.
Zurück zum Zitat Goldstein I, Levy T, Galili D, Ovadia H, Yirmiya R, Rosen H, et al. Involvement of Na(+), K(+)-ATPase and endogenous digitalis-like compounds in depressive disorders. Biol Psychiatry. 2006;60:491–9.CrossRefPubMed Goldstein I, Levy T, Galili D, Ovadia H, Yirmiya R, Rosen H, et al. Involvement of Na(+), K(+)-ATPase and endogenous digitalis-like compounds in depressive disorders. Biol Psychiatry. 2006;60:491–9.CrossRefPubMed
60.
Zurück zum Zitat DeGiorgis JA, Jaffe H, Moreira JE, Carlotti CG, Leite JP, Pant HC, et al. Phosphoproteomic analysis of synaptosomes from human cerebral cortex. J Proteome Res. 2005;4:306–15.CrossRefPubMed DeGiorgis JA, Jaffe H, Moreira JE, Carlotti CG, Leite JP, Pant HC, et al. Phosphoproteomic analysis of synaptosomes from human cerebral cortex. J Proteome Res. 2005;4:306–15.CrossRefPubMed
61.
Zurück zum Zitat Dieterich DC, Kreutz MR. Proteomics of the synapse—a quantitative approach to neuronal plasticity. Mol Cell Proteom. 2016;15:368–81.CrossRef Dieterich DC, Kreutz MR. Proteomics of the synapse—a quantitative approach to neuronal plasticity. Mol Cell Proteom. 2016;15:368–81.CrossRef
62.
Zurück zum Zitat Zhang Y, Guo Z, Zou L, Yang Y, Zhang L, Ji N, et al. Data for a comprehensive map and functional annotation of the human cerebrospinal fluid proteome. Data Brief. 2015;3:103–7.CrossRefPubMedPubMedCentral Zhang Y, Guo Z, Zou L, Yang Y, Zhang L, Ji N, et al. Data for a comprehensive map and functional annotation of the human cerebrospinal fluid proteome. Data Brief. 2015;3:103–7.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Nanjappa V, Thomas JK, Marimuthu A, Muthusamy B, Radhakrishnan A, Sharma R, et al. Plasma proteome database as a resource for proteomics research: 2014 update. Nucleic Acids Res. 2014;42:D959–65.CrossRefPubMed Nanjappa V, Thomas JK, Marimuthu A, Muthusamy B, Radhakrishnan A, Sharma R, et al. Plasma proteome database as a resource for proteomics research: 2014 update. Nucleic Acids Res. 2014;42:D959–65.CrossRefPubMed
64.
Zurück zum Zitat Chiu MJ, Yang SY, Horng HE, Yang CC, Chen TF, Chieh JJ, et al. Combined plasma biomarkers for diagnosing mild cognition impairment and Alzheimer’s disease. ACS Chem Neurosci. 2013;4:1530–6.CrossRefPubMedPubMedCentral Chiu MJ, Yang SY, Horng HE, Yang CC, Chen TF, Chieh JJ, et al. Combined plasma biomarkers for diagnosing mild cognition impairment and Alzheimer’s disease. ACS Chem Neurosci. 2013;4:1530–6.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Cirrito JR, Yamada KA, Finn MB, Sloviter RS, Bales KR, May PC, et al. Synaptic activity regulates interstitial fluid amyloid-β levels in vivo. Neuron. 2005;48:913–22.CrossRefPubMed Cirrito JR, Yamada KA, Finn MB, Sloviter RS, Bales KR, May PC, et al. Synaptic activity regulates interstitial fluid amyloid-β levels in vivo. Neuron. 2005;48:913–22.CrossRefPubMed
66.
Zurück zum Zitat Sokolow S, Henkins KM, Bilousova T, Gonzalez B, Vinters HV, Miller CA, et al. Pre-synaptic C-terminal truncated tau is released from cortical synapses in Alzheimer’s disease. J Neurochem. 2015;133:368–79.CrossRefPubMedPubMedCentral Sokolow S, Henkins KM, Bilousova T, Gonzalez B, Vinters HV, Miller CA, et al. Pre-synaptic C-terminal truncated tau is released from cortical synapses in Alzheimer’s disease. J Neurochem. 2015;133:368–79.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Gylys KH, Fein JA, Yang F, Cole GM. Enrichment of presynaptic and postsynaptic markers by size-based gating analysis of synaptosome preparations from rat and human cortex. Cytom A. 2004;60:90–6.CrossRef Gylys KH, Fein JA, Yang F, Cole GM. Enrichment of presynaptic and postsynaptic markers by size-based gating analysis of synaptosome preparations from rat and human cortex. Cytom A. 2004;60:90–6.CrossRef
68.
Zurück zum Zitat Sokolow S, Henkins KM, Williams IA, Vinters HV, Schmid I, Cole GM, et al. Isolation of synaptic terminals from Alzheimer’s disease cortex. Cytom A. 2012;81:248–54.CrossRef Sokolow S, Henkins KM, Williams IA, Vinters HV, Schmid I, Cole GM, et al. Isolation of synaptic terminals from Alzheimer’s disease cortex. Cytom A. 2012;81:248–54.CrossRef
69.
Zurück zum Zitat Gylys KH, Fein JA, Yang F, Wiley DJ, Miller CA, Cole GM. Synaptic changes in Alzheimer’s disease: increased amyloid-beta and gliosis in surviving terminals is accompanied by decreased PSD-95 fluorescence. Am J Pathol. 2004;165:1809–17.CrossRefPubMedPubMedCentral Gylys KH, Fein JA, Yang F, Wiley DJ, Miller CA, Cole GM. Synaptic changes in Alzheimer’s disease: increased amyloid-beta and gliosis in surviving terminals is accompanied by decreased PSD-95 fluorescence. Am J Pathol. 2004;165:1809–17.CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat Gylys KH, Fein JA, Yang F, Miller CA, Cole GM. Increased cholesterol in Abeta-positive nerve terminals from Alzheimer’s disease cortex. Neurobiol Aging. 2007;28:8–17.CrossRefPubMed Gylys KH, Fein JA, Yang F, Miller CA, Cole GM. Increased cholesterol in Abeta-positive nerve terminals from Alzheimer’s disease cortex. Neurobiol Aging. 2007;28:8–17.CrossRefPubMed
71.
Zurück zum Zitat Fein JA, Sokolow S, Miller CA, Vinters HV, Yang F, Cole GM, et al. Co-localization of amyloid beta and tau pathology in Alzheimer’s disease synaptosomes. Am J Pathol. 2008;172:1683–92.CrossRefPubMedPubMedCentral Fein JA, Sokolow S, Miller CA, Vinters HV, Yang F, Cole GM, et al. Co-localization of amyloid beta and tau pathology in Alzheimer’s disease synaptosomes. Am J Pathol. 2008;172:1683–92.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Bilousova T, Miller CA, Poon WW, Vinters HV, Corrada M, Kawas C, et al. Synaptic amyloid-beta oligomers precede p-tau and differentiate high pathology control cases. Am J Pathol. 2016;186:185–98.CrossRefPubMedPubMedCentral Bilousova T, Miller CA, Poon WW, Vinters HV, Corrada M, Kawas C, et al. Synaptic amyloid-beta oligomers precede p-tau and differentiate high pathology control cases. Am J Pathol. 2016;186:185–98.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Postupna NO, Keene CD, Latimer C, Sherfield EE, Van Gelder RD, Ojemann JG, et al. Flow cytometry analysis of synaptosomes from post-mortem human brain reveals changes specific to Lewy body and Alzheimer’s disease. Lab Invest. 2014;94:1161–72.CrossRefPubMedPubMedCentral Postupna NO, Keene CD, Latimer C, Sherfield EE, Van Gelder RD, Ojemann JG, et al. Flow cytometry analysis of synaptosomes from post-mortem human brain reveals changes specific to Lewy body and Alzheimer’s disease. Lab Invest. 2014;94:1161–72.CrossRefPubMedPubMedCentral
74.
Zurück zum Zitat Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci. 2010;11:155–9.PubMed Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci. 2010;11:155–9.PubMed
75.
Zurück zum Zitat Walker LC, Diamond MI, Duff KE, Hyman BT. Mechanisms of protein seeding in neurodegenerative diseases. JAMA Neurol. 2013;70:304–10.CrossRefPubMed Walker LC, Diamond MI, Duff KE, Hyman BT. Mechanisms of protein seeding in neurodegenerative diseases. JAMA Neurol. 2013;70:304–10.CrossRefPubMed
76.
Zurück zum Zitat Daniel JA, Malladi CS, Kettle E, McCluskey A, Robinson PJ. Analysis of synaptic vesicle endocytosis in synaptosomes by high-content screening. Nat Protoc. 2012;7:1439–55.CrossRefPubMed Daniel JA, Malladi CS, Kettle E, McCluskey A, Robinson PJ. Analysis of synaptic vesicle endocytosis in synaptosomes by high-content screening. Nat Protoc. 2012;7:1439–55.CrossRefPubMed
77.
Zurück zum Zitat Heilemann M, Van De Linde S, Schüttpelz M, Kasper R, Seefeldt B, Mukherjee A, et al. Subdiffraction-resolution fluorescence imaging with conventional fluorescent probes. Angew Chem Int Ed. 2008;47:6172–6.CrossRef Heilemann M, Van De Linde S, Schüttpelz M, Kasper R, Seefeldt B, Mukherjee A, et al. Subdiffraction-resolution fluorescence imaging with conventional fluorescent probes. Angew Chem Int Ed. 2008;47:6172–6.CrossRef
78.
79.
Zurück zum Zitat DeKosky ST, Scheff SW. Synapse loss in frontal cortex biopsies in Alzheimer’s disease: correlation with cognitive severity. Ann Neurol. 1990;27:457–64.CrossRefPubMed DeKosky ST, Scheff SW. Synapse loss in frontal cortex biopsies in Alzheimer’s disease: correlation with cognitive severity. Ann Neurol. 1990;27:457–64.CrossRefPubMed
80.
Zurück zum Zitat Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R, et al. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991;30:572–80.CrossRefPubMed Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R, et al. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991;30:572–80.CrossRefPubMed
81.
Zurück zum Zitat Arold S, Sullivan P, Bilousova T, Teng E, Miller CA, Poon WW, et al. Apolipoprotein E level and cholesterol are associated with reduced synaptic amyloid beta in Alzheimer’s disease and apoE TR mouse cortex. Acta Neuropathol. 2012;123:39–52.CrossRefPubMed Arold S, Sullivan P, Bilousova T, Teng E, Miller CA, Poon WW, et al. Apolipoprotein E level and cholesterol are associated with reduced synaptic amyloid beta in Alzheimer’s disease and apoE TR mouse cortex. Acta Neuropathol. 2012;123:39–52.CrossRefPubMed
82.
Zurück zum Zitat Henkins KM, Sokolow S, Miller CA, Vinters HV, Poon WW, Cornwell LB, et al. Extensive p-tau pathology and SDS-stable p-tau oligomers in Alzheimer’s cortical synapses. Brain Pathol. 2012;22:826–33.CrossRefPubMedPubMedCentral Henkins KM, Sokolow S, Miller CA, Vinters HV, Poon WW, Cornwell LB, et al. Extensive p-tau pathology and SDS-stable p-tau oligomers in Alzheimer’s cortical synapses. Brain Pathol. 2012;22:826–33.CrossRefPubMedPubMedCentral
83.
Zurück zum Zitat Koffie RM, Hashimoto T, Tai HC, Kay KR, Serrano-Pozo A, Joyner D, et al. Apolipoprotein E4 effects in Alzheimer’s disease are mediated by synaptotoxic oligomeric amyloid-β. Brain. 2012;135:2155–68.CrossRefPubMedPubMedCentral Koffie RM, Hashimoto T, Tai HC, Kay KR, Serrano-Pozo A, Joyner D, et al. Apolipoprotein E4 effects in Alzheimer’s disease are mediated by synaptotoxic oligomeric amyloid-β. Brain. 2012;135:2155–68.CrossRefPubMedPubMedCentral
84.
Zurück zum Zitat Ahmed Z, Cooper J, Murray TK, Garn K, McNaughton E, Clarke H, et al. A novel in vivo model of tau propagation with rapid and progressive neurofibrillary tangle pathology: the pattern of spread is determined by connectivity, not proximity. Acta Neuropathol. 2014;127:667–83.CrossRefPubMedPubMedCentral Ahmed Z, Cooper J, Murray TK, Garn K, McNaughton E, Clarke H, et al. A novel in vivo model of tau propagation with rapid and progressive neurofibrillary tangle pathology: the pattern of spread is determined by connectivity, not proximity. Acta Neuropathol. 2014;127:667–83.CrossRefPubMedPubMedCentral
85.
Zurück zum Zitat de Calignon A, Polydoro M, Suarez-Calvet M, William C, Adamowicz DH, Kopeikina KJ, et al. Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron. 2012;73:685–97.CrossRefPubMedPubMedCentral de Calignon A, Polydoro M, Suarez-Calvet M, William C, Adamowicz DH, Kopeikina KJ, et al. Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron. 2012;73:685–97.CrossRefPubMedPubMedCentral
86.
Zurück zum Zitat Dujardin S, Lecolle K, Caillierez R, Begard S, Zommer N, Lachaud C, et al. Neuron-to-neuron wild-type Tau protein transfer through a trans-synaptic mechanism: relevance to sporadic tauopathies. Acta Neuropathol Commun. 2014;2:14.CrossRefPubMedPubMedCentral Dujardin S, Lecolle K, Caillierez R, Begard S, Zommer N, Lachaud C, et al. Neuron-to-neuron wild-type Tau protein transfer through a trans-synaptic mechanism: relevance to sporadic tauopathies. Acta Neuropathol Commun. 2014;2:14.CrossRefPubMedPubMedCentral
87.
88.
Zurück zum Zitat Pooler AM, Polydoro M, Maury EA, Nicholls SB, Reddy SM, Wegmann S, et al. Amyloid accelerates tau propagation and toxicity in a model of early Alzheimer’s disease. Acta Neuropathol Commun. 2015;3:14.CrossRefPubMedPubMedCentral Pooler AM, Polydoro M, Maury EA, Nicholls SB, Reddy SM, Wegmann S, et al. Amyloid accelerates tau propagation and toxicity in a model of early Alzheimer’s disease. Acta Neuropathol Commun. 2015;3:14.CrossRefPubMedPubMedCentral
89.
Zurück zum Zitat Perez-Nievas BG, Stein TD, Tai HC, Dols-Icardo O, Scotton TC, Barroeta-Espar I, et al. Dissecting phenotypic traits linked to human resilience to Alzheimer’s pathology. Brain. 2013;136:2510–26.CrossRefPubMedPubMedCentral Perez-Nievas BG, Stein TD, Tai HC, Dols-Icardo O, Scotton TC, Barroeta-Espar I, et al. Dissecting phenotypic traits linked to human resilience to Alzheimer’s pathology. Brain. 2013;136:2510–26.CrossRefPubMedPubMedCentral
90.
Zurück zum Zitat Tan CC, Yu JT, Wang HF, Tan MS, Meng XF, Wang C, et al. Efficacy and safety of donepezil, galantamine, rivastigmine, and memantine for the treatment of Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimers Dis. 2014;41:615–31.PubMed Tan CC, Yu JT, Wang HF, Tan MS, Meng XF, Wang C, et al. Efficacy and safety of donepezil, galantamine, rivastigmine, and memantine for the treatment of Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimers Dis. 2014;41:615–31.PubMed
91.
Zurück zum Zitat Whitehouse PJ, Price DL, Clark AW, Coyle JT, DeLong MR. Alzheimer disease: evidence for selective loss of cholinergic neurons in the nucleus basalis. Ann Neurol. 1981;10:122–6.CrossRefPubMed Whitehouse PJ, Price DL, Clark AW, Coyle JT, DeLong MR. Alzheimer disease: evidence for selective loss of cholinergic neurons in the nucleus basalis. Ann Neurol. 1981;10:122–6.CrossRefPubMed
92.
Zurück zum Zitat Whitehouse PJ, Price DL, Struble RG, Clark AW, Coyle JT, Delon MR. Alzheimer’s disease and senile dementia: loss of neurons in the basal forebrain. Science. 1982;215:1237.CrossRefPubMed Whitehouse PJ, Price DL, Struble RG, Clark AW, Coyle JT, Delon MR. Alzheimer’s disease and senile dementia: loss of neurons in the basal forebrain. Science. 1982;215:1237.CrossRefPubMed
Metadaten
Titel
The Study of Postmortem Human Synaptosomes for Understanding Alzheimer’s Disease and Other Neurological Disorders: A Review
verfasst von
Jia-Fong Jhou
Hwan-Ching Tai
Publikationsdatum
01.07.2017
Verlag
Springer Healthcare
Erschienen in
Neurology and Therapy / Ausgabe Sonderheft 1/2017
Print ISSN: 2193-8253
Elektronische ISSN: 2193-6536
DOI
https://doi.org/10.1007/s40120-017-0070-z

Weitere Artikel der Sonderheft 1/2017

Neurology and Therapy 1/2017 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.