Skip to main content
American Journal of Neurodegenerative Disease logoLink to American Journal of Neurodegenerative Disease
. 2012 May 15;1(1):32–48.

Can zebrafish be used as animal model to study Alzheimer's disease?

Soraya Santana 1, Eduardo P Rico 2, Javier S Burgos 1
PMCID: PMC3560447  PMID: 23383380

Abstract

Zebrafish is rapidly emerging as a promising model organism to study various central nervous system (CNS) disorders, including Alzheimer’s disease (AD). AD is the main cause of dementia in the human population and there is an urgency to understand the causes of this neurodegenerative disease. In this respect, the development of new animal models to study the underlying neurodegenerative mechanisms of AD is an urgent need. In this review we analyze the current situation in the use of zebrafish as a model for AD, discussing the reasons to use this experimental paradigm in CNS investigation and analyzing the several strategies adopted to induce an AD-like pathology in zebrafish. We discuss the strategies of performing interventions to cause damage in the zebrafish brain by altering the major neurotransmitter systems (such as cholinergic, glutamatergic or GABAergic circuits). We also analyze the several transgenic zebrafish constructed for the AD study, discussing both the familial-AD models based on APP processing pathway (APP and presenilins) and in the TAU hyperphosphorylation, together with the genes involved in sporadic-AD, as apolipoprotein E. We conclude that zebrafish is in a preliminary stage of development in the AD field, and that the transgenic animals must be improved to use this fish as an optimal model for AD research. Furthermore, a deeper knowledge of the zebrafish brain and a better characterization of the injury caused by alterations in the major neurotransmitter systems are needed.

Keywords: zebrafish, Alzheimer’s disease, neurotoxins, cholinergic neurotransmission, glutamatergic neurotransmission, GABAergic neurotransmission, apolipoprotein E, APP, presenilin, TAU

1. Alzheimer’s disease (AD)

Alzheimer’s disease (AD) is the most common form of dementia characterized mainly by massive neuronal loss and impaired synaptic processes located in the cerebral cortex, particularly in the frontal and temporal lobes, and hippocampus [1]. For over a century, pathologic features have been used to define AD, whose phenotype is composed of two main hallmarks: (i) the deposition of senile plaques, which are extracellular amyloid-β (Aβ) peptide aggregates with axons and dendrites usually swollen or atrophic, and (ii) the neurofibrillary tangles composed of intracellular hyperphosphorylated TAU protein and aggregated into paired helical filaments [1].

Despite the intense research devoted to the elucidation of the mechanisms underlying AD, these are so far unknown. It is only well-documented the cause of a percentage less than 1% of cases, known as familial AD (FAD), which shows a pattern of autosomal dominant inheritance and early onset of symptoms. FAD is a monogenic disease associated with the presence of specific mutations mainly in three genes that increase the production of Aβ peptide of 42 amino acids [2]: amyloid-β precursor protein (APP), presenilin 1 (PSEN1), and to a lesser extent the presenilin 2 (PSEN2). The rest of the cases (>99%), called sporadic AD (SAD), presents a complex interaction between genetic susceptibility factors and environmental being the main risk factor for AD, the allele ε4 of the gene for apolipoprotein E (APOEε4) [3].

2. Zebrafish as a model for AD

Historically, rodent models have been used to study AD. However, the causes of AD remain unknown, and more efforts to decipher the mechanisms underlying neurodegeneration are necessary. Despite the progress in this field, we still need a better understanding of AD, which supports the growing importance of further innovative research using experimental models of neurodegeneration. Therefore, additional animal models with complementary advantages must be used to analyze the basis of the neurodegeneration and, subsequently, to evaluate the effects of novel drugs, as a previous step to assays in rodents. In this respect, the zebrafish (Danio rerio) have recently become a focus of neurobehavioral studies since it displays neuropathological and behavioural phenotypes that are quantifiable [4,5], and it has been recently proposed as a valid experimental paradigm to study AD [6]. Zebrafish is an ideal model for the study of human diseases because they present a number of features that make it unique as an animal model. For this reason, the zebrafish have experienced a dramatic rise in popularity as an experimental organism.

Zebrafish offer a reasonable compromise between physiological complexity and throughput, have a fully characterized genome, and display significant physiological homology to mammals, including humans [7]. Moreover, it presents a clear advantage in comparison to other animal models: the larva-adult duality. The availability of both forms is beneficial and enables the investigation of a wider spectrum of neurodegenerative-related phenomena throughout ontogenesis. The faster development and longer lifespan of zebrafish, compared to mice, makes them and ideal choice to model developmental trajectories of neurodegeneration. Moreover, as rodent models are expensive to maintain and more difficult to modify genetically, lower organisms emerge as useful species to increase the knowledge of the mechanisms underlying neurodegenerative processes. The zebrafish is a vertebrate more closely related to humans than invertebrate models such as yeast, worms or flies, which design and connectivity in the central nervous system (CNS) correlates with the human [8]. Although invertebrates can provide important insights into neurodegeneration, the absence of a complex nervous system limits their application in modelling intricate aspects of CNS disorders. Comparison between various species is also important for uncovering mechanisms associated to brain pathogenesis. While the complementary use of zebrafish could be an important strategy for AD research, it represents an integral part of a more global cross-species modelling (from fish to rodents) for uncovering evolutionary conserved mechanisms of neurodegeneration.

Furthermore, zebrafish has a short time of development to sexual maturity (3 months) and a high reproductive rate (hundreds of embryos per female per clutch and per week). Embryos are transparent and developed externally, facts that allow direct observation of embryogenesis and development of the CNS. Embryos are also easily amenable to methods for manipulating genes and protein activity such as injection of antisense oligonucleotides [9,10], mRNAs or transgenes, and for screening of drug libraries being arrayed in microtitre plates [11]. Moreover, whereas the early zebrafish developmental stages are likely to be important to analyze CNS processes and abnormalities, adult fish with the full range of brain functions should have a significant advantage in the analysis of the complex brain functions characteristics of vertebrates [12], and in the studies of conductual phenotyping. Indeed, zebrafish exhibit many higher order behaviors including memory, conditioned responses, and social behaviors like schooling [13]. All these properties strengthen the zebrafish as an ideal model for studying human diseases, including CNS disorders [14].

3. The zebrafish brain organization

Zebrafish has been recently recognized as a reliable model for studies of behaviour, neural circuitry and neural disease [8], due to its phylogenetical proximity to the human. The comparison of the zebrafish brain structure with man shows a high conservation of basic brain organization [15], together with similar key neuroanatomical [16,17] and neurochemical [18] pathways of relevance to human diseases. The organization of the zebrafish brain is similar to other vertebrates, despite the smaller cerebral hemispheres and the structure and function of the optic tectum, having similarly defined areas such as the hypothalamus and olfactory bulb, encompassing structures of the lateral pallium (located in the telencephalon), which appear to be homologous to the mammalian hippocampus. Indeed, zebrafish and mammalian encephalon share many structural properties, such as the main organization (fore-, mid- and hind-brain, including diencephalon, telencephalon and cerebellum), or the principal neurotransmitter systems [18]. The major neurotransmitter circuits, such as the glutamatergic (excitatory) and the GABAergic (inhibitory), have been demonstrated to exist in the zebrafish brain [5]. In addition to the major excitatory and inhibitory neurotransmitters, muscarinic cholinergic receptors have been confirmed in brain extracts of adult zebrafish by radioligand binding methods [19]. Moreover, the neurotransmitters such as GABA, dopamine, serotonin, histamine, glutamate and acetylcholine are also found in zebrafish [12].

Additionally, zebrafish CNS also contains the main cellular types found in the mammalian brain, such as microglia [20], oligodendrocytes and myelin [21,22], cerebellar Purkinje cells [23], motor neurons [24] or astrocytes [25].

This species also possess a tight junction-based blood-brain barrier, with substantial macromolecule permeability, and highly regulated [26], a fact essential to the search for novel neuroprotective compounds [27].

3.1. The zebrafish cholinergic neurotransmission

Acetylcholine (ACh) is an essential neurotransmitter in the central and peripheral nervous system (PNS) that developed very early in phylogenetical history. The cholinergic system performs a critical role in learning processes and memory functions, wherein their projections are severely affected in AD [28]. Indeed, AD is related to cholinergic system dysfunctions such as the loss of cholinergic neurons in the basal forebrain and hippocampus [29,30], and in this respect, memantine (a glutamate receptor NMDA antagonist) has been currently used for the treatment of AD [31].

The presence of a cholinergic system in the zebrafish brain has been verified through electrophysiological, histological, and biochemical approaches. Electrophysiological studies demonstrated that carbachol, a muscarinic receptor agonist, modulated the electrically-induced neural activity in the isolated zebrafish brain [19]. Histologically, neurons containing acetylcholinesterase (AChE) were detected in most areas of the zebrafish CNS, including the olfactory bulb, telencephalon, cerebellum, medulla oblongata, and spinal cord [32]. In particular, the telencephalon, which is thought to be responsible for learning in teleost fish [33,34], appeared to have AChE-positive neurons in various nuclei of the dorsal and ventral areas. The identification of cholinergic neurons in zebrafish CNS has been also previously reported by using specific antibodies against choline acetyltransferase (ChAT) [18,32,35]. In particular, the anatomical identification of ChAT immunoreactive neurons in adult zebrafish differs among the studies probably due to the distinct methodological approaches. In any case, ChAT appears to be present in the ventral and dorsal telencephalic areas [18], wherein the presence of ChAT immunoreactive axons indicates that the AChE-positive neurons are cholinoceptive. Finally, the main developmental pattern of ChAT positive neurons have been described for zebrafish [36].

Therefore, the cholinergic system of zebrafish has been shown to be widely distributed, according to the results of the immunocytochemical studies on ChAT and AChE in the zebrafish brain.

3.2. The zebrafish glutamatergic neurotransmission

Glutamate is the most widespread and important excitatory neurotransmitter in the CNS of vertebrates. However, an overstimulation of the glutamatergic system may lead to excitotoxicity, a phenomenon involved in many pathological conditions including AD [37,38]

The neurotransmitter glutamate activates several classes of metabotropic receptor and three major types of ionotropic receptors: 2-amino-3-(5-methyl-3-oxo-1,2- oxazol-4-yl) propanoic acid (AMPA), kainate (KA) and N-methyl-D-aspartate (NMDA) [39]. The molecular characterization and embryonic expression of the family of NMDA receptor subunit genes has already been established in zebrafish [40]. Considering that long-term potentiation is the representative synaptic modification underlying the process of learning and memory, Nam and collaborators [41] studied the NMDA receptor-dependent long-term potentiation in the telencephalon of the zebrafish, which is analogous to hippocampus and amygdala of mammalian brain [33,34]. In this context, a simple protocol of inhibitory avoidance task in adult zebrafish demonstrated that the resulting memory is robust, long-lasting and sensitive to NMDA-receptor antagonist MK-801 given in the tank water immediately after the training session [42]. Furthermore, a biochemical study showed that MK-801 alter the Na+, K+ -ATPase activity and oxidative parameters in zebrafish brain [43].

Moreover Edwards and Michel [44] demonstrated the pharmacological characterization of ionotropic glutamatergic receptors in olfactory bulb in zebrafish. This group advanced the understanding of the glutamatergic system in teleosts by characterizing the distribution of functional NMDA and KA-stimulated neurons. Furthermore, Tabor and Friedrich [45] pharmacologically investigated the ionotropic glutamate receptor function in neuronal circuits of the zebrafish olfactory bulb.

Other via to neutralize the excess of extracellular glutamate is the high-affinity uptake mechanism executed by a family of excitatory amino acid transporters (EAATs) [37]. The presence of EAAT-related sequences in zebrafish CNS have been recently described [46]. Once identified these EAAT-related genes, some preliminary parameters of glutamate transporter activity were investigated through sodium-dependent glutamate uptake in distinct zebrafish brain structures [46].

3.3. The zebrafish GABAergic neurotransmission

The γ-aminobutyric acid (GABA) is a classical inhibitory neurotransmitter in the brain, playing essential roles in synaptic plasticity [47]. Defects in GABAergic neurotransmission are the cause of several CNS pathologies [48,49]. In addition, given the hypothesis that the abusive stimulation of ionotropic glutamate receptors is in part responsible for the pathology of AD, it is important to note that disruptions or alterations in GABA-mediated neurotransmission may significantly impact the functional relationship between excitatory and inhibitory systems [50,51]. In fact, the balance between excitatory and inhibitory neurotransmitters is involved in several events in CNS, with many neurodegenerative diseases being correlated with brain cells dysfunction, and, consequently, causing changes in these signaling systems.

GABA-containing neurons appear in the zebrafish olfactory bulb, telencephalon, tectum stratum, and in the hypothalamus [52]. In the cerebellar corpus and valves of the zebrafish, GABA receptors are present in the molecular layer, Purkinje cells, and groups of Golgi cells in the granular layer [53]. Furthermore, an electrophysiological study demonstrated the evidence of GABAA and GABAC receptors on zebrafish retinal bipolar cells [54]. Accordingly, immunohistochemical studies found expression of GABAergic cells during early embryonic stages in the zebrafish brain, and an increase in their number with developmental progression [55].

Glutamic acid decarboxylase (GAD) catalyzes the α-decarboxylation of glutamic acid to produce γ-aminobutyric acid. GAD is highly conserved and has been described in Drosophila, avian, and mammalian species [56,57]. Martin and collaborators [58] analyzed the amino acid identities and similarities between zebrafish and human GAD65 and GAD67 cDNAs. Moreover, in situ hybridization revealed that both GAD65 and GAD67 were expressed early in the zebrafish embryo during the period of axonogenesis, suggesting a role for GABA prior to synapse formation. Both GAD genes were detected in the telencephalon, in the nucleus of the medial longitudinal fasciculus in the midbrain, and at the border regions of the rhombomeres in the rostral hindbrain.

Based on these data, the evaluation of parameters associated to GABAergic system emerges as a tempting strategy to assess neurochemical, behavioral, and toxicological phenotypes associated to AD-related models in both larval and adult zebrafish.

4. Pharmacological models for AD in zebrafish

4.1. Cholinergic neurotoxins

AD is characterized by the functional decreased of the cholinergic system, which is crucial to cognitive impairment. The early expression of AChE in diverse cell types suggests that it may have a developmental role and thus, could be a putative target for neurotoxicity in zebrafish [59]. The first study that used biochemical and molecular approaches demonstrated the enzyme activity and gene expression pattern for AChE in zebrafish brain [60]. Successive reports addressed the importance of this enzyme in regulating zebrafish brain functions. Afterwards, the effects of several drugs on cholinergic transmission have been evaluated. In this regard, the effects promoted by metals [61-63], typical and atypical antipsychotics [43], antiepileptics [64] and ethanol [65] on AChE activity have already been studied in zebrafish brain.

To demonstrate the activity of the cholinergic system, the effects induced by scopolamine (a cholinergic muscarinic receptor antagonist) have been analyzed in zebrafish. Similarly to what occurs on mammals, the results showed that this drug impairs both the acquisition of passive avoidance response and retention of the learned response, and that physostigmine (an acetylcholinesterase inhibitor that blocks the breakdown of the released ACh at the synaptic site) rescues the amnesic effects of scopolamine [66] (Table 1). In this regard, the use of both quercetin and rutin prevented scopolamine-induced memory impairment in zebrafish [63]. These results are very consistent with similar studies in rodents showing scopolamine-induced learning deficits in rats and mice [67-69].

Table 1.

Summary of recent studies of pharmacological models for AD utilizing larval and adult zebrafish (MOA: mechanism of action).

Neurotransmitter system Neurotoxin MOA Model Endpoint Reference
Cholinergic Scopolamine Muscarinic receptor antagonist Adults Behavior [63,66]
Carbachol Muscarinic receptor agonist Adults Electrophysiology [19]
Pilocarpine Muscarinic receptor agonist Adults Behavior [72]
    Larvae Locomotor activity [71]
Physostigmine Acetylcholinesterase inhibitor Adults Behavior [73]

Glutamatergic MK-801 NMDA-receptor antagonist Adults Behavior [77]
Ketamine NMDA-receptor antagonist Adults Behavior & Molecular biology [88-90]
APV NMDA-receptor antagonist Adults Electrophysiology [41]
Memantine NMDA-receptor antagonist Larvae Behavior [84,85]
Kainate KA receptor agonist Adults Behavior [80]
Domoate KA receptor agonist Larvae Behavior [81]
CNQX AMPA-receptor antagonist Adults Electrophysiology [41]

GABAergic Pentylentetrazole GABA-receptor antagonsit Larvae Behavior, electrophysiology, molecular biology & biochemistry [83,93,97]
    Adults Behavior [73,95,96]
Picrotoxin GABA-receptor antagonsit Adults Behavior [95]
Phenylpyrazole GABA-receptor antagonist Larvae Development [91]
Valproate GABA transaminase, voltage-gated sodium channels & T-type calcium channels inhibitor Adults Behavior, electrophysiology, molecular biology [96]

In addition, saturation binding experiments showed relatively high receptor numbers and affinity for cholinergic agonists and antagonists including oxotremorine, carbachol, atropine, and pirenzepine [70]. In this respect, two of these muscarinic receptor agonists (carbachol and oxotremorine) suppressed the electrically evoked field potentials in the telencephalon of the adult zebrafish in a reversible and dose-dependent manner [19].

Another muscarinic receptor agonist, pilocarpine, is frequently used as neurotoxic drug inducing seizures. In fact, the anticonvulsant and/or proconvulsant effects of antidepressants against pilocarpine- and pentylenetetrazole-induced seizures in larval zebrafish and mice has been determined [71]. Additionally, the cholinergic effects promoted by nicotine and pilocarpine on dopaminergic system have been verified in neurobehavioral parameters induced by chlorpyrifos exposure [72], and inhibitors of AChE such as soman and physostigmine also induce seizures, which further cause an excessive release of excitatory amino acids [73].

Altogether, these findings could facilitate the use of the zebrafish as a model for study of cholinergic mechanisms underlying learning and memory.

4.2. Glutamatergic neurotoxins

Glutamate-mediated neurotoxicity, also known as excitotoxicity, is associated to the AD pathogenesis, being caused by the over activation of the glutamate receptors, that ultimately leads to neuronal damage/death [74]. Given the early loss of glutamatergic neurons in AD in vulnerable pathways such as the medial temporal lobe/hippocampal network, the role of excitotoxic mechanisms has long been associated to this disease [75]. In this respect, memantine (a low potency NMDA receptor antagonist) is the main blockbuster drug for AD (together with cholinesterase inhibitors), although only a clinically poor symptomatic benefit has been demonstrated [76].

To date, the research in the field of glutamatergic transmission in adult zebrafish has been confined to studies analyzing the effects of MK-801 on their swimming behaviour, learning and memory [77] and to histological descriptions of ionotropic glutamate receptors [44] (Table 1). The presence of NMDA- and KA-sensitive ionotropic glutamate receptors was immunohistochemically detected in the zebrafish olfactory bulb [78]. Electrophysiologically, neural activity was blocked by CNQX, an AMPA receptor antagonist, and d-2-amino-5-phosphono-valerate (APV), an NMDA receptor antagonist, in the telencephalon [41]. In this respect, blockade of NMDA receptor activation that is crucial in the learning process in rats also blocks acquisition of passive avoidance learning in zebrafish [42]. Moreover, the synaptic plasticity induced in the medial division of zebrafish telencephalon was inhibited by CNQX [79].

More recently, we have demonstrate that KA receptors in zebrafish operate in a similar way to those in established models, wherein the KA-induced seizures can be specifically inhibited in a dose-dependent fashion by DNQX [80]. The inhibition of seizures by specific glutamate receptor antagonists makes the zebrafish a discriminatory model to study the excitotoxicity, even though a demonstration of a specific neuronal death after excitotoxicity in adult zebrafish is still remaining. These results are in line with previous reports using domoic acid (DA) in larval zebrafish [81]. DA, a structural relative to kainate and the neurotransmitter glutamate, activates AMPA and kainate subtypes of the glutamate receptor family resulting in excitotoxicity predominantly in brain tissues [82]. The microinjection of DA to fertilized eggs induced toxicity on zebrafish development [81]. These toxics effects include reduced hatching and uncontrolled pectoral fin motions and tonic-clonic like convulsions [81]. Moreover, in ovo exposure to DA increases the susceptibility of larval zebrafish to the seizure-inducing agent PTZ [83].

The effect of memantine has been also studied in the zebrafish model. Best and co-workers demonstrated that zebrafish larvae exhibit iterative reduction in a startle response to a series of acoustic stimuli, with memantine increasing this acoustic startle response and decreased habituation [84]. On the other hand, exposure to memantine did not alter the swim speed of zebrafish larvae [85].

Finally, ketamine, a non-competitive antagonist of NMDA receptor, has been used to induce sedation and analgesia [86,87]. Sub-anesthetic doses of ketamine produced a variety of abnormal behaviors, such as altered gill movement, stress responses and aberrant circling behavior in adult zebrafish [88]. Acute exposure of ketamine reduced anxiety, impaired intra-session habituation and evoked circular swimming of adult zebrafish. Additionally, ketamine reduced whole-body cortisol levels and elevated brain c-fos expression in zebrafish [89]. Finally, Kanungo and collaborators demonstrated that ketamine adversely affected motor neuron axon length and decreased cranial and motor neuron populations [90].

Considering the involvement of the glutamatergic system in the AD pathogenesis, and that the results obtained with glutamatergic neurotoxins in zebrafish and rodents are qualitatively comparable [81,85], it becomes important to consider the zebrafish as a model for future developments in AD.

4.3. GABAergic neurotoxins

The investigation of environmental toxins (e.g.,organic compounds and heavy metals) has been used to understand the zebrafish GABAergic neurotransmission. The use of phenylpyrazole insecticide (fipronil) on embryos showed that, although this insecticide acts as an inhibitor of GABA receptors, it may also inhibit a structurally related glycine receptor subtype expressed during the development of spinal locomotor pathways [91] (Table 1).

On the other hand, pentylenetetrazole (PTZ) acts via blockade of the GABAergic system, in particular by inhibiting the GABAA receptor-mediated inhibitory postsynaptic potentials [92]. In zebrafish, exposure to PTZ induced a concentration-dependent sequence of behavioral changes culminating in clonus-like convulsions [93], that can be inhibited by common antiepileptic drugs, such as valproate or diazepam, and different agents that block synaptic transmission, like tetrodotoxin (to block Na+-dependent action potentials), kynurenate (a non-specific blocker of postsynaptic glutamate receptors) and a cocktail containing non-NMDA and NMDA glutamate receptors blockers [93]. Additionally, PTZ-treated adult zebrafish showed deficits in the acquisition and maintenance of passive avoidance response [73]. Furthermore, there is evidence about epilepsy and a disorder of GABA metabolism through succinic semialdehyde dehidrogenase deficiency [94]. Moreover, it has been described that PTZ and picrotoxin evoke seizure-like states were evoked in adult zebrafish [95].

These impairments in the learning of passive avoidance responses after PTZ treatment are suppressed with a valproate pretreatment [96]. More recently, it has been reported that low doses of PTZ can reverse the normal behavioral response to alternating periods of light and dark in zebrafish larvae [97].

These results, together, indicate that the GABAergic system can be impaired in zebrafish, being a relevant target for the study of the balance between neuronal circuits, and for the evaluation of antiepileptic drugs and neuroprotectants.

4.4. AD neurotoxins

The classical neurotoxins used in rodents to mimic the AD pathology have not yet been developed in zebrafish. Although the pharmacological disruption of Aβ clearance has been proposed as of great interest in AD, only one study of Aβ administration in larval zebrafish has been documented [98]. The administration of Aβ40 induced premature senescence in the vascular endothelium, impairing angiogenesis [98]. Moreover, although the effects of exogenously applied Aβ have been widely examined in rodents [99], in adult zebrafish no effect of Aβ administration has been developed or investigated to date.

Because hyperphosphorylated and aggregated forms of TAU protein are present in AD brain and cerebrospinal fluid, much recent attention has been devoted to investigating TAU in animal models [100,101]. Experimentally, TAU hyperphosphorylation has been mimicked by okadaic acid (OA) in culture cells [102] and in animals [103]. OA is a potent inhibitor of phosphatases 1 and 2A, that induces characteristics that resemble AD-like pathology such as memory impairment accompanied by remarkable neuropathological changes including hippocampal neurodegeneration, a paired helical filament-like phosphorylation of tau protein, formation of β-amyloid containing plaque-like structures, oxidative stress and specific astroglial alterations [103,104]. To the best of our knowledge, OA has not been evaluated in the zebrafish.

5. Transgenic zebrafish for AD

One important advantage of the zebrafish is the genome organization and the genetic pathways controlling signal transduction and development, because they are highly conserved between zebrafish and vertebrates [105]. Indeed, the zebrafish genes share as well as 50-80% homology with most human sequences and, in this respect, several genome databases similar to mouse have been generated: the Zebrafish Informartion Network (ZFIN) [106] that contains mounting genetic and phenotypic data, or the recently developed Zebrafish Neurophenome Project (ZNP) database [107] that collects the repository of neurobehavioral and related physiological phenotypes in zebrafish. For these reasons, transgenic zebrafish are continuously increasing in number and being used to model human diseases.

5.1. APP pathway

5.1.1. APP

There are two main routes of APP protein processing; (i) the predominant one is the non-amyloidogenic route and involves the enzyme called α-secretase, that excludes the generation of β-amyloid peptide [108]; (ii) the other is the amyloidogenic pathway, in which Aβ peptide is generated by the sequential action of two proteases known as β- and γ-secretase [109]. This route mainly generates two species of peptide Aβ: Aβ40 (of 40 amino acids) and Aβ42 (of 42). Although the most abundant isoform is Aβ40 (approximately 90%) the insoluble Aβ42 peptide has a greater tendency to aggregate into fibrils and therefore is more cytotoxic than the Aβ40 peptide [110].

Zebrafish have two genes homologues of human APP gene, appa and appb [111] (Table 2). The gene appa encodes a protein that is about 70% identical to the one encoded by appb. The human APP695 and zebrafish appa have approximately 70% amino acid identity, with 80% identity in the Aβ42 region, 95% identity within the transmembrane domain and 94% identity in the cytoplasmic region. The Aβ region within appb protein is 71% identical to human Aβ, 94% identical in the cytoplasmic region and 100% identical in the transmembrane region [111]. Appb more closely resembles human APP695 that is expressed exclusively in the mammalian brain, while the presence of the Kunitz Protease inhibiotr (KPI) domain makes appa more similar to the human APP770 isoform, which is expressed ubiquitously [112]. Expression of appa and appb is widespread throughout different developmental stages. These two genes show differences in their expression patterns during the segmentation period, whereas at 24 hours post-fertilization (hpf), appa and appb shared anterior expression in the telencephalon, the ventral diencephalon, the trigeminal ganglia and the posterior lateral line ganglia [111].

Table 2.

Genes involved in sporadic and familial AD, the homologous genes in zebrafish and the percentage of protein sequence homology.

Human Gene Zebrafish gene Protein homology Reference
APP appa 70% [111]
appb 70%
PSEN1 psen1 74% [118]
PSEN2 psen2 74% [119]
Pen2 Pen2 74% [129]
Aph1A&B Aph1 62% [129]
Nicastrin Nicastrin 56% [128]
TAU mapta/maptb Unidentified [135]
APOE APOE 28% [143]

Regarding to APP zebrafish models, Lee and coworkers used the zebrafish appb promoter to express GFP in transgenic animals [113] (Table 3). This model revealed that appb is mainly expressed in sub regions of brain, spinal cord and the developing vasculature of the zebrafish embryos. Moreover, in adult transgenic zebrafish, appb was abundantly expressed in the brain (telencephalon, optic tectum, thalamus and cerebellum) [113]. The demonstration that zebrafish appb gene regulatory elements can be used to drive GFP expression in both embryos and adults will be important for future studies that focus on expressing mutant human APP in zebrafish. In this study, the authors predicted that zebrafish expressing mutant human APP will generate Aβ plaques, although did not demonstrate their presence.

Table 3.

Zebrafish transgenic/knockdown models for AD (CNS: Central Nervous System. MO: morpholino).

Human Gene Zebrafish gene Protein homology Reference
APP appb promoter- GFP appb expression in larvae and adults [113]
appa and appb MO Reduced body weight & defective movements [114]
PSEN1 psen1 MO Somite defects & defective brain development [120,121]
PSEN2 psen2 MO Altered gene expression & CNS defects [123,124]
TAU Transient overexpression of 4R-Tau-GFP TAU phosphorylation & tangle-like structures [137]
Human 4R/0N tau TAU accumulation resembling tangles [136]
Human TAU-P301L TAU hyperphosphorylation, tangles & neuronal death [138]

In other study, Joshi and co-workers knocked down the zebrafish appa and appb function by morpholino (MO) injection resulting in a shortened body axis, a short, curly tail, mild synopthalmia and defective convergent-extension movements during gastrulation [114]. Injecting appa-MO showed no or mild defects in developing embryos whereas appb-MO caused phenotypic changes similar to those observed when both MOs were injected together. These defects can be rescued by human APP695 mRNA, partially rescued by human APPsα mRNA, but cannot be rescued by APP bearing the Swedish mutation (APPswe) [114]. These data suggest that APP processing plays an important role in APP function and that APPswe is functionally deficient and the possibility that APPswe may not only contribute to AD pathogenesis during aging but also may exert changes during embryonic development.

5.1.2. Presenilins

Presenilins (PSENs) are the most well-known proteins belonging to the catalytic subunit of the proteolytic γ-secretase complex, that cleave Notch and APP [115]. Two separate PSEN genes have been identified in humans, PSEN1 and PSEN2, which show a high sequence homology. PSEN1 and PSEN2 are membrane spanning proteins, which are endoproteolytically cleaved into two stable fragments, one N-terminal and other C-terminal. Models of familial AD mutations have demonstrated an enhancement of APP proteolytic processing through aberrations of the γ-secretase complex [116]. These alterations generally increase the production of Aβ42 or decrease the production of Aβ40, which results in changes to the Aβ40/Aβ42 ratio [117].

Orthologues of human presenilin genes, psen1 [118] and psen2 [119] have been identified in zebrafish, which show a high degree of sequence identity to the human PSEN1 and PSEN2, respectively (Table 2). Sequence comparison to human PSEN1 demonstrates that the protein encoded by the zebrafish psen1 is 74% identical to PSEN1. Expression of psen1 was detected in blastomeres before the onset of zygotic transcription, demonstrating maternal inheritance. Similar to human PSEN1, zebrafish presenilin-1 requires two critical aspartate residues for its function, and overexpression of zebrafish presenilin-1 in human cells promotes increased Aβ production [118].

In zebrafish, reduction of psen1 activity by MO injection leads to disruption of somite formation consistent with partial loss of Notch signaling in the presomitic mesoderm [120] (Table 3). Moreover, embryos injected with the psen1 MO have a shorter tail, and a smaller head. Reduction of psen1 results in the loss of Notch target gene her6 (orthologue of human HES1) expression and increased expression of neurogenin 1 (ngn1) mRNA, a marker of neural progenitor cells, further indicative of reduced Notch signaling [121].

The zebrafish gene psen2 encodes a protein that is about 74% identical to the one encoded by human PSEN2 and 70% amino acids identity to the zebrafish psen1 [122] (Table 2). Alignment of the zebrafish psen2 and human PSEN2 protein sequences shows that the primary structure is highly conserved between them with only the N-terminal and the C-terminal fragments of the cytoplasmic loop domain being highly variable. [119]. Zebrafish psen2 is ubiquitously expressed during early embryogenesis, but becomes more restricted by 24 hpf. Psen2 protein expression is initiated between 6 and 12 hpf, suggesting strict regulation of psen2 translation. At 24 hpf, high levels of psen2 transcript could be detected in the anterior developing CNS and eye, in the midline of the developing spinal cord, and also in cells derived from the neural crest [119].

In zebrafish, reducing psen2 expression by MO injection give rise a phenotype resembling loss of psen1 activity, included hydrocephalus and reduced pigmentation [123,124] (Table 3). Moreover, blockade of translation of zebrafish psen2 transcripts produces significant changes in embryo development including expansion of brain ventricles at 48 hpf and decreased trunk neural crest formation with increased numbers of interneurons and dorsal sensory neurons at 24 hpf [124]. A combined decrease of psen1 and psen2 activity partially reverses the effect of psen2 MO, suggesting an opposing effect of psen1 and psen2, pointing a non-redundant roles for both presenilins in zebrafish development [124].

5.1.3. Other genes of the APP pathway

Besides the PSENs, other important proteins of the γ-secretase complex are nicastrin, anterior pharynx defective 1 (APH-1), and presenilin enhancer 2 (PEN-2) [125]. Nicastrin is a glycosylated integral membrane protein that binds to both the N- and C-terminal fragments of PSENs. Suppressing nicastrin expression using siRNA results in decreased levels of PSENs, indicating that nicastrin is one of the stabilizing factors of the PSENs fragments [126]. These proteins stabilize each other upon assembly and allow for the correct trafficking of the mature complex to the cell surface and to endocytic compartments where γ-secretase exerts its activity [127].

APH-1, PEN-2 and nicastrin have been identified in zebrafish. The gene nicastrin has been identified and has sequenced in zebrafish by several groups, showing a 56% homology to human nicastrin [128] (Table 2). Zebrafish pen-2 has 74% homology to human PEN-2, and zebrafish aph-1 has 62% residues that are identical to both human homologues of APH-1 (APH-1A and APH-1B) [129]. Both zebrafish genes, Pen-2 and Aph-1, are present from the fertilization, being ubiquitously expressed in the embryo at 12 hpf and, predominantly in the brain at 24 hpf [121].

However, despite conservation of the Aβ domain and of the secretases between zebrafish and humans, a zebrafish Aβ peptide has not yet been found and it is not known if the above post-translational modifications that occur in human APP processing also occur in zebrafish.

5.2. TAU

Microtubule-associated protein TAU (MAPT) plays a large role in the outgrowth of neuronal processes and the development of neuronal polarity. TAU promotes microtubule assembly, stabilizes microtubules, and affects the dynamics of microtubules in neurons [130], being abundantly present in the CNS and predominantly expressed in neuronal axons [131]. The phosphorylation of TAU regulates microtubule binding and assembly [132]. In contrast, pathological TAU becomes hyperphosphorylated, which destabilizes microtubules by decreased binding to microtubules, resulting in the aggregation of hyperphosphorylated TAU. TAU dysfunction is a major histological characteristic of AD and is common factor in a number of other neurodegenerative disorders included frontotemporal dementia with Parkinson linked to chromosome 17 (FTDP-17), corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP) [133].

Recent developments strengthen the zebrafish as a model for the study of tauopathies (reviewed by [134]). In this regard, two paralogues of MAPT in zebrafish, called mapta and maptb has been identified (Table 2). The two proteins encoded by the zebrafish genes have not yet been detected, but both mapta and maptb mRNAs were expressed in the developing CNS [135]. A complex pattern of alternative splicing of the mapta and maptb transcripts suggests that, like human TAU, zebrafish tau isoforms with different numbers of microtubule-binding repeats are expressed in the CNS, and larger forms of tau are expressed in the PNS [135]. Moreover, the zebrafish genome contains highly conserved orthologues of each of the kinases implicated in tau phosphorylation [136].

Regarding the TAU zebrafish models, Tomasiewicz and collaborators developed a fish transiently expressing mutant human TAU fused to GFP under the control of the zebrafish neural-specific GATA-2 promoter [137] (Table 3). GFP-positive neurons were found in the brain, retina and spinal cord. This mutant form of human TAU expressed in zebrafish neurons produced a cytoskeletal disruption that closely resembled the neurofibrillary tangles in human disease. Further, the human TAU-GFP fusion was phosphorylated in the zebrafish brain demonstrating that human TAU could be a substrate for kinases expressed in the larval zebrafish brain.

Afterwards, Bai and co-workers developed a stable transgenic zebrafish line expressing the 4-repeat isoform of the human TAU under transcriptional control of the eno2 promoter [136]. This stable transgenic Tg(eno2:Tau) zebrafish showed widespread CNS neuronal expression of TAU, at high levels compared with normal human brain. Human 4R-Tau was found within axons, neuropil and in ectopic accumulations in neuronal cell bodies of Tg(eno2:Tau) zebrafish.

More recently, a transgenic zebrafish line expressing the human protein TAU-P301L (a mutation genetically linked to frontotemporal dementia) in neurons by the Gal4-upstream activating-based (Gal4/UAS-based) vector system has been generated [138]. This model allows monitoring the early pathology, including disease-specific hyperphosphorylation and conformational changes of TAU, neuronal and behavioral abnormalities. Finally, the use of inhibitors of human GSK3β reduced TAU phosphorylation in this transgenic zebrafish, suggesting that this model can be used for the search of drugs that inhibit human TAU phosphorylation.

5.3. APOE

To date, the main genetic risk factor associated with the sporadic form of AD is the allele 4 of the apolipoprotein E gene (APOE-ε4) [3,139]. Presence of one copy of the APOE-ε4 allele increases the risk to suffer AD by about three times, while two copies increase by about 12 times [139]. Furthermore, the presence of one or two copies of APOE-ε4 lead to an earlier age of onset by about 10-20 years compared with non-carriers [3]. However, the apoE allele-ε4 is neither necessary nor sufficient for disease development [140]. For this reason, although there have been numerous studies attempting to elucidate how the APOE influences AD, the underlying mechanisms of this association have not yet been deciphered [141].

A zebrafish gene homologous to mammalian APOE has been previously identified [142] (Table 2). However, deduced amino acid sequences of apoE shows only a 28% of identity with human apoE sequence [143], and a different genomic organization. The zebrafish gene consists of five exons (an additional intron that splits the last exon in two exons) instead of four in the mammalian gene. Although the homology between zebrafish apoE and human APOE is low, a region in zebrafish apoE enriched in basic amino acids is similar to the lipoprotein receptor binding domain of human apoE. ApoE is expressed in the zebrafish eyes, and some cells of the mesencephalic, telencephalic, and rhombencephalic brain areas, suggesting that it may play a significant function in the CNS [143]. Moreover, a complex APOE expression pattern has been demonstrated during embryonic and larval development of zebrafish [143,144]. Functionally apoE seems to be important for zebrafish nutrition since it is expressed in the yolk syncytial layer [143], suggesting that zebrafish can be used as a simple and useful model for studying the role of apoE in brain morphogenesis and regeneration. However, to the best of our knowledge, APOE zebrafish transgenic models have not been developed to date.

6. Conclusions

In the present review, we highlight the main advances that zebrafish offers in order to understand the pathological mechanisms of Alzheimer’s disease (AD). Here, we reinforce the idea that this vertebrate is an interesting tool that can be strategically incorporated into the analysis of the neurodegeneration cascade, covering the existing gap between the drug discovery in cellular models and the preclinical assays in rodents. In any case, zebrafish should be used in large scale before pharmacological validation in rodent models, especially because of its larva-adult duality. In fact, over the last years, zebrafish has emerged as an attractive model for AD-related research.

Although we have provided diverse examples to demonstrate the scope for zebrafish to model AD, several pieces of the puzzle are still lacking. For example, a better understanding of the comparative brain anatomy and physiology of adult zebrafish will be required, and more transgenic zebrafish to model an AD-like pathology are a current need. Indeed, the most important gene to build a valid AD zebrafish model is the APOE, because of this apolipoprotein is the main risk factor for AD, after aging.

Moreover, few pharmacological models causing specific neurotoxicity have been developed to date. In the future, we will need fast models of neuronal damage, in a similar way than the mouse models based in the neurotoxin injections have been historically developed. In this sense, Aβ and TAU modulation experimental systems will be needed to decipher the mechanisms of AD pathology in zebrafish. In any case, to date, and in a similar way than the wild-type mouse models, Aβ deposits have not been found in the zebrafish brain.

In addition, a higher knowledge of the specific neuronanatomical circuits participating in the neurodegeneration must be elucidated, especially those highly related with the mammalian hippocampus, such as the zebrafish telencephalon.

We concluded that a more effort must be done to definitely optimize the zebrafish as a valid model for AD. Afterwards, the complementary use of several animal models including zebrafish, with distinct pros and contras, will help us to understand the molecular basis of the AD, and to develop novel strategies to prevent the neurodegenerative diseases.

Acknowledgments

The authors thank Professor Fernando Valdivieso for his continuous encouragement and help, and for his critical reading of this manuscript. We also acknowledge the critical commentaries of Dr. Jose Luis Gómez-Skarmeta.

This work was funded by the financial resources of Neuron Bio and by grants from the Agencia de Innovación y Desarrollo de Andalucía, the CDTI of the Ministerio de Ciencia e Innovación, and by funds from the Torres Quevedo program (SS and JSB).

EPR was a recipient of fellowship from DECIT/SCTIE-MS through Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Fundação de Amparo à Pesquisa do Estado do Rio Grande do Sul (FAPERGS) (Proc. 10/0036-5, Conv n. 700545/2008), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) and by the FINEP research grant “Rede Instituto Brasileiro de Neurociência (IBN-Net)” # 01.06.0842-00.

Disclosures

SS and JSB have served as paid researchers for Neuron Bio.

References

  • 1.Blennow K, de Leon MJ, Zetterberg H. Alzheimer's disease. Lancet. 2006;368:387–403. doi: 10.1016/S0140-6736(06)69113-7. [DOI] [PubMed] [Google Scholar]
  • 2.Scheuner D, Eckman C, Jensen M, Song X, Citron M, Suzuki N, Bird TD, Hardy J, Hutton M, Kukull W, Larson E, Levy-Lahad E, Viitanen M, Peskind E, Poorkaj P, Schellenberg G, Tanzi R, Wasco W, Lannfelt L, Selkoe D, Younkin S. Secreted amyloid beta-protein similar to that in the senile plaques of Alzheimer's disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer's disease. Nat Med. 1996;2:864–870. doi: 10.1038/nm0896-864. [DOI] [PubMed] [Google Scholar]
  • 3.Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, Roses AD, Haines JL, Pericak-Vance MA. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science. 1993;261:921–923. doi: 10.1126/science.8346443. [DOI] [PubMed] [Google Scholar]
  • 4.Bowman TV, Zon LI. Swimming into the future of drug discovery: in vivo chemical screens in zebrafish. ACS Chem Biol. 2010;5:159–161. doi: 10.1021/cb100029t. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rico EP, Rosemberg DB, Seibt KJ, Capiotti KM, Da Silva RS, Bonan CD. Zebrafish neurotransmitter systems as potential pharmacological and toxicological targets. Neurotoxicol Teratol. 2011;33:608–617. doi: 10.1016/j.ntt.2011.07.007. [DOI] [PubMed] [Google Scholar]
  • 6.Newman M, Verdile G, Martins RN, Lardelli M. Zebrafish as a tool in Alzheimer's disease research. Biochim Biophys Acta. 2011;1812:346–352. doi: 10.1016/j.bbadis.2010.09.012. [DOI] [PubMed] [Google Scholar]
  • 7.Barbazuk WB, Korf I, Kadavi C, Heyen J, Tate S, Wun E, Bedell JA, McPherson JD, Johnson SL. The syntenic relationship of the zebrafish and human genomes. Genome Res. 2000;10:1351–1358. doi: 10.1101/gr.144700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Guo S. Linking genes to brain, behavior and neurological diseases: what can we learn from zebrafish? Genes Brain Behav. 2004;3:63–74. doi: 10.1046/j.1601-183x.2003.00053.x. [DOI] [PubMed] [Google Scholar]
  • 9.Nasevicius A, Ekker SC. Effective targeted gene 'knockdown' in zebrafish. Nat Genet. 2000;26:216–220. doi: 10.1038/79951. [DOI] [PubMed] [Google Scholar]
  • 10.Eisen JS, Smith JC. Controlling morpholino experiments: don't stop making antisense. Development. 2008;135:1735–1743. doi: 10.1242/dev.001115. [DOI] [PubMed] [Google Scholar]
  • 11.Zon LI, Peterson RT. In vivo drug discovery in the zebrafish. Nat Rev Drug Discov. 2005;4:35–44. doi: 10.1038/nrd1606. [DOI] [PubMed] [Google Scholar]
  • 12.Panula P, Sallinen V, Sundvik M, Kolehmainen J, Torkko V, Tiittula A, Moshnyakov M, Podlasz P. Modulatory neurotransmitter systems and behavior: towards zebrafish models of neurodegenerative diseases. Zebrafish. 2006;3:235–247. doi: 10.1089/zeb.2006.3.235. [DOI] [PubMed] [Google Scholar]
  • 13.Lieschke GJ, Currie PD. Animal models of human disease: zebrafish swim into view. Nat Rev Genet. 2007;8:353–367. doi: 10.1038/nrg2091. [DOI] [PubMed] [Google Scholar]
  • 14.Grunwald DJ, Eisen JS. Headwaters of the zebrafish -- emergence of a new model vertebrate. Nat Rev Genet. 2002;3:717–724. doi: 10.1038/nrg892. [DOI] [PubMed] [Google Scholar]
  • 15.Wullimann MF, Rupp B, Reichert H. Neuroanatomy of the Zebrafish Brain. Berlín: Birkhauser-Verlag; 1996. [Google Scholar]
  • 16.Mueller T, Wullimann MF. An evolutionary interpretation of teleostean forebrain anatomy. Brain Behav Evol. 2009;74:30–42. doi: 10.1159/000229011. [DOI] [PubMed] [Google Scholar]
  • 17.Rink E, Wullimann MF. Connections of the ventral telencephalon (subpallium) in the zebrafish (Danio rerio) Brain Res. 2004;1011:206–220. doi: 10.1016/j.brainres.2004.03.027. [DOI] [PubMed] [Google Scholar]
  • 18.Mueller T, Vernier P, Wullimann MF. The adult central nervous cholinergic system of a neurogenetic model animal, the zebrafish Danio rerio. Brain Res. 2004;1011:156–169. doi: 10.1016/j.brainres.2004.02.073. [DOI] [PubMed] [Google Scholar]
  • 19.Park E, Lee Y, Kim Y, Lee CJ. Cholinergic modulation of neural activity in the telencephalon of the zebrafish. Neurosci Lett. 2008;439:79–83. doi: 10.1016/j.neulet.2008.04.064. [DOI] [PubMed] [Google Scholar]
  • 20.Peri F, Nusslein-Volhard C. Live imaging of neuronal degradation by microglia reveals a role for v0-ATPase a1 in phagosomal fusion in vivo. Cell. 2008;133:916–927. doi: 10.1016/j.cell.2008.04.037. [DOI] [PubMed] [Google Scholar]
  • 21.Avila RL, Tevlin BR, Lees JP, Inouye H, Kirschner DA. Myelin structure and composition in zebrafish. Neurochem Res. 2007;32:197–209. doi: 10.1007/s11064-006-9136-5. [DOI] [PubMed] [Google Scholar]
  • 22.Yoshida M, Macklin WB. Oligodendrocyte development and myelination in GFP-transgenic zebrafish. J Neurosci Res. 2005;81:1–8. doi: 10.1002/jnr.20516. [DOI] [PubMed] [Google Scholar]
  • 23.Koulen P, Janowitz T, Johnston LD, Ehrlich BE. Conservation of localization patterns of IP (3) receptor type 1 in cerebellar Purkinje cells across vertebrate species. J Neurosci Res. 2000;61:493–499. doi: 10.1002/1097-4547(20000901)61:5<493::AID-JNR3>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
  • 24.Westerfield M, McMurray JV, Eisen JS. Identified motoneurons and their innervation of axial muscles in the zebrafish. J Neurosci. 1986;6:2267–2277. doi: 10.1523/JNEUROSCI.06-08-02267.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kawai H, Arata N, Nakayasu H. Three-dimensional distribution of astrocytes in zebrafish spinal cord. Glia. 2001;36:406–413. doi: 10.1002/glia.1126. [DOI] [PubMed] [Google Scholar]
  • 26.Jeong JY, Kwon HB, Ahn JC, Kang D, Kwon SH, Park JA, Kim KW. Functional and developmental analysis of the blood-brain barrier in zebrafish. Brain Res Bull. 2008;75:619–628. doi: 10.1016/j.brainresbull.2007.10.043. [DOI] [PubMed] [Google Scholar]
  • 27.Eliceiri BP, Gonzalez AM, Baird A. Zebrafish model of the blood-brain barrier: morphological and permeability studies. Methods Mol Biol. 2011;686:371–378. doi: 10.1007/978-1-60761-938-3_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bierer LM, Haroutunian V, Gabriel S, Knott PJ, Carlin LS, Purohit DP, Perl DP, Schmeidler J, Kanof P, Davis KL. Neurochemical correlates of dementia severity in Alzheimer's disease: relative importance of the cholinergic deficits. J Neurochem. 1995;64:749–760. doi: 10.1046/j.1471-4159.1995.64020749.x. [DOI] [PubMed] [Google Scholar]
  • 29.Auld DS, Kornecook TJ, Bastianetto S, Quirion R. Alzheimer's disease and the basal forebrain cholinergic system: relations to beta-amyloid peptides, cognition, and treatment strategies. Prog Neurobiol. 2002;68:209–245. doi: 10.1016/s0301-0082(02)00079-5. [DOI] [PubMed] [Google Scholar]
  • 30.Schliebs R. Basal forebrain cholinergic dysfunction in Alzheimer's disease--interrelationship with beta-amyloid, inflammation and neurotrophin signaling. Neurochem Res. 2005;30:895–908. doi: 10.1007/s11064-005-6962-9. [DOI] [PubMed] [Google Scholar]
  • 31.Howard R, McShane R, Lindesay J, Ritchie C, Baldwin A, Barber R, Burns A, Dening T, Findlay D, Holmes C, Hughes A, Jacoby R, Jones R, McKeith I, Macharouthu A, O'Brien J, Passmore P, Sheehan B, Juszczak E, Katona C, Hills R, Knapp M, Ballard C, Brown R, Banerjee S, Onions C, Griffin M, Adams J, Gray R, Johnson T, Bentham P, Phillips P. Donepezil and memantine for moderate-to-severe Alzheimer's disease. N Engl J Med. 2012;366:893–903. doi: 10.1056/NEJMoa1106668. [DOI] [PubMed] [Google Scholar]
  • 32.Clemente D, Porteros A, Weruaga E, Alonso JR, Arenzana FJ, Aijon J, Arevalo R. Cholinergic elements in the zebrafish central nervous system: Histochemical and immunohistochemical analysis. J Comp Neurol. 2004;474:75–107. doi: 10.1002/cne.20111. [DOI] [PubMed] [Google Scholar]
  • 33.Portavella M, Vargas JP, Torres B, Salas C. The effects of telencephalic pallial lesions on spatial, temporal, and emotional learning in goldfish. Brain Res Bull. 2002;57:397–399. doi: 10.1016/s0361-9230(01)00699-2. [DOI] [PubMed] [Google Scholar]
  • 34.Rodriguez F, Lopez JC, Vargas JP, Broglio C, Gomez Y, Salas C. Spatial memory and hippocampal pallium through vertebrate evolution: insights from reptiles and teleost fish. Brain Res Bull. 2002;57:499–503. doi: 10.1016/s0361-9230(01)00682-7. [DOI] [PubMed] [Google Scholar]
  • 35.Kaslin J, Nystedt JM, Ostergard M, Peitsaro N, Panula P. The orexin/hypocretin system in zebrafish is connected to the aminergic and cholinergic systems. J Neurosci. 2004;24:2678–2689. doi: 10.1523/JNEUROSCI.4908-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Arenzana FJ, Clemente D, Sanchez-Gonzalez R, Porteros A, Aijon J, Arevalo R. Development of the cholinergic system in the brain and retina of the zebrafish. Brain Res Bull. 2005;66:421–425. doi: 10.1016/j.brainresbull.2005.03.006. [DOI] [PubMed] [Google Scholar]
  • 37.Danbolt NC. Glutamate uptake. Prog Neurobiol. 2001;65:1–105. doi: 10.1016/s0301-0082(00)00067-8. [DOI] [PubMed] [Google Scholar]
  • 38.Maragakis NJ, Rothstein JD. Glutamate transporters: animal models to neurologic disease. Neurobiol Dis. 2004;15:461–473. doi: 10.1016/j.nbd.2003.12.007. [DOI] [PubMed] [Google Scholar]
  • 39.Hu NW, Ondrejcak T, Rowan MJ. Glutamate receptors in preclinical research on Alzheimer's disease: update on recent advances. Pharmacol Biochem Behav. 2012;100:855–862. doi: 10.1016/j.pbb.2011.04.013. [DOI] [PubMed] [Google Scholar]
  • 40.Cox JA, Kucenas S, Voigt MM. Molecular characterization and embryonic expression of the family of N-methyl-D-aspartate receptor subunit genes in the zebrafish. Dev Dyn. 2005;234:756–766. doi: 10.1002/dvdy.20532. [DOI] [PubMed] [Google Scholar]
  • 41.Nam RH, Kim W, Lee CJ. NMDA receptor-dependent long-term potentiation in the telencephalon of the zebrafish. Neurosci Lett. 2004;370:248–251. doi: 10.1016/j.neulet.2004.08.037. [DOI] [PubMed] [Google Scholar]
  • 42.Blank M, Guerim LD, Cordeiro RF, Vianna MR. A one-trial inhibitory avoidance task to zebrafish: rapid acquisition of an NMDA-dependent long-term memory. Neurobiol Learn Mem. 2009;92:529–534. doi: 10.1016/j.nlm.2009.07.001. [DOI] [PubMed] [Google Scholar]
  • 43.Seibt KJ, da Luz Oliveira R, Rosemberg DB, Savio LE, Scherer EB, Schmitz F, Wyse AT, Bonan CD. MK-801 alters Na(+), K (+)-ATPase activity and oxidative status in zebrafish brain: reversal by antipsychotic drugs. J Neural Transm. 2011 doi: 10.1007/s00702-011-0745-z. [DOI] [PubMed] [Google Scholar]
  • 44.Edwards JG, Michel WC. Pharmacological characterization of ionotropic glutamate receptors in the zebrafish olfactory bulb. Neuroscience. 2003;122:1037–1047. doi: 10.1016/s0306-4522(03)00544-x. [DOI] [PubMed] [Google Scholar]
  • 45.Tabor R, Friedrich RW. Pharmacological analysis of ionotropic glutamate receptor function in neuronal circuits of the zebrafish olfactory bulb. PLoS One. 2008;3:e1416. doi: 10.1371/journal.pone.0001416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rico EP, de Oliveira DL, Rosemberg DB, Mussulini BH, Bonan CD, Dias RD, Wofchuk S, Souza DO, Bogo MR. Expression and functional analysis of Na(+)-dependent glutamate transporters from zebrafish brain. Brain Res Bull. 2010;81:517–523. doi: 10.1016/j.brainresbull.2009.11.011. [DOI] [PubMed] [Google Scholar]
  • 47.Staley KJ, Soldo BL, Proctor WR. Ionic mechanisms of neuronal excitation by inhibitory GABAA receptors. Science. 1995;269:977–981. doi: 10.1126/science.7638623. [DOI] [PubMed] [Google Scholar]
  • 48.Luddens H, Korpi ER, Seeburg PH. GABAA/benzodiazepine receptor heterogeneity: neurophysiological implications. Neuropharmacology. 1995;34:245–254. doi: 10.1016/0028-3908(94)00158-o. [DOI] [PubMed] [Google Scholar]
  • 49.Mori H, Mishina M. Structure and function of the NMDA receptor channel. Neuropharmacology. 1995;34:1219–1237. doi: 10.1016/0028-3908(95)00109-j. [DOI] [PubMed] [Google Scholar]
  • 50.Rissman RA, De Blas AL, Armstrong DM. GABA(A) receptors in aging and Alzheimer's disease. J Neurochem. 2007;103:1285–1292. doi: 10.1111/j.1471-4159.2007.04832.x. [DOI] [PubMed] [Google Scholar]
  • 51.Lanctot KL, Herrmann N, Mazzotta P, Khan LR, Ingber N. GABAergic function in Alzheimer's disease: evidence for dysfunction and potential as a therapeutic target for the treatment of behavioural and psychological symptoms of dementia. Can J Psychiatry. 2004;49:439–453. doi: 10.1177/070674370404900705. [DOI] [PubMed] [Google Scholar]
  • 52.Kim YJ, Nam RH, Yoo YM, Lee CJ. Identification and functional evidence of GABAergic neurons in parts of the brain of adult zebrafish (Danio rerio) Neurosci Lett. 2004;355:29–32. doi: 10.1016/j.neulet.2003.10.024. [DOI] [PubMed] [Google Scholar]
  • 53.Delgado L, Schmachtenberg O. Immunohistochemical localization of GABA, GAD65, and the receptor subunits GABAAalpha1 and GABAB1 in the zebrafish cerebellum. Cerebellum. 2008;7:444–450. doi: 10.1007/s12311-008-0047-7. [DOI] [PubMed] [Google Scholar]
  • 54.Connaughton VP, Nelson R, Bender AM. Electrophysiological evidence of GABAA and GABAC receptors on zebrafish retinal bipolar cells. Vis Neurosci. 2008;25:139–153. doi: 10.1017/S0952523808080322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Doldan MJ, Prego B, Holmqvist BI, de Miguel E. Distribution of GABA-immunolabeling in the early zebrafish (Danio rerio) brain. Eur J Morphol. 1999;37:126–129. doi: 10.1076/ejom.37.2.126.4748. [DOI] [PubMed] [Google Scholar]
  • 56.Jackson FR, Newby LM, Kulkarni SJ. Drosophila GABAergic systems: sequence and expression of glutamic acid decarboxylase. J Neurochem. 1990;54:1068–1078. doi: 10.1111/j.1471-4159.1990.tb02359.x. [DOI] [PubMed] [Google Scholar]
  • 57.Erlander MG, Tillakaratne NJ, Feldblum S, Patel N, Tobin AJ. Two genes encode distinct glutamate decarboxylases. Neuron. 1991;7:91–100. doi: 10.1016/0896-6273(91)90077-d. [DOI] [PubMed] [Google Scholar]
  • 58.Martin SC, Heinrich G, Sandell JH. Sequence and expression of glutamic acid decarboxylase isoforms in the developing zebrafish. J Comp Neurol. 1998;396:253–266. [PubMed] [Google Scholar]
  • 59.Hanneman E, Westerfield M. Early expression of acetylcholinesterase activity in functionally distinct neurons of the zebrafish. J Comp Neurol. 1989;284:350–361. doi: 10.1002/cne.902840303. [DOI] [PubMed] [Google Scholar]
  • 60.Rico EP, Rosemberg DB, Senger MR, Arizi Mde B, Bernardi GF, Dias RD, Bogo MR, Bonan CD. Methanol alters ecto-nucleotidases and acetylcholinesterase in zebrafish brain. Neurotoxicol Teratol. 2006;28:489–496. doi: 10.1016/j.ntt.2006.05.001. [DOI] [PubMed] [Google Scholar]
  • 61.Senger MR, Rico EP, de Bem Arizi M, Frazzon AP, Dias RD, Bogo MR, Bonan CD. Exposure to Hg2+ and Pb2+ changes NTPDase and ecto-5'-nucleotidase activities in central nervous system of zebrafish (Danio rerio) Toxicology. 2006;226:229–237. doi: 10.1016/j.tox.2006.07.012. [DOI] [PubMed] [Google Scholar]
  • 62.Senger MR, Rosemberg DB, Rico EP, de Bem Arizi M, Dias RD, Bogo MR, Bonan CD. In vitro effect of zinc and cadmium on acetylcholinesterase and ectonucleotidase activities in zebrafish (Danio rerio) brain. Toxicol In Vitro. 2006;20:954–958. doi: 10.1016/j.tiv.2005.12.002. [DOI] [PubMed] [Google Scholar]
  • 63.Richetti SK, Blank M, Capiotti KM, Piato AL, Bogo MR, Vianna MR, Bonan CD. Quercetin and rutin prevent scopolamine-induced memory impairment in zebrafish. Behav Brain Res. 2011;217:10–15. doi: 10.1016/j.bbr.2010.09.027. [DOI] [PubMed] [Google Scholar]
  • 64.Siebel AM, Rico EP, Capiotti KM, Piato AL, Cusinato CT, Franco TM, Bogo MR, Bonan CD. In vitro effects of antiepileptic drugs on acetylcholinesterase and ectonucleotidase activities in zebrafish (Danio rerio) brain. Toxicol In Vitro. 2010;24:1279–1284. doi: 10.1016/j.tiv.2010.03.018. [DOI] [PubMed] [Google Scholar]
  • 65.Rico EP, Rosemberg DB, Dias RD, Bogo MR, Bonan CD. Ethanol alters acetylcholinesterase activity and gene expression in zebrafish brain. Toxicol Lett. 2007;174:25–30. doi: 10.1016/j.toxlet.2007.08.005. [DOI] [PubMed] [Google Scholar]
  • 66.Kim YH, Lee Y, Kim D, Jung MW, Lee CJ. Scopolamine-induced learning impairment reversed by physostigmine in zebrafish. Neurosci Res. 2010;67:156–161. doi: 10.1016/j.neures.2010.03.003. [DOI] [PubMed] [Google Scholar]
  • 67.Janas AM, Cunningham SC, Duffy KB, Devan BD, Greig NH, Holloway HW, Yu QS, Markowska AL, Ingram DK, Spangler EL. The cholinesterase inhibitor, phenserine, improves Morris water maze performance of scopolamine-treated rats. Life Sci. 2005;76:1073–1081. doi: 10.1016/j.lfs.2004.06.028. [DOI] [PubMed] [Google Scholar]
  • 68.Xu J, Rong S, Xie B, Sun Z, Zhang L, Wu H, Yao P, Zhang Y, Liu L. Procyanidins extracted from the lotus seedpod ameliorate scopolamine-induced memory impairment in mice. Phytother Res. 2009;23:1742–1747. doi: 10.1002/ptr.2837. [DOI] [PubMed] [Google Scholar]
  • 69.Eidi M, Zarrindast MR, Eidi A, Oryan S, Parivar K. Effects of histamine and cholinergic systems on memory retention of passive avoidance learning in rats. Eur J Pharmacol. 2003;465:91–96. doi: 10.1016/s0014-2999(03)01440-7. [DOI] [PubMed] [Google Scholar]
  • 70.Williams FE, Messer WS Jr. Muscarinic acetylcholine receptors in the brain of the zebrafish (Danio rerio) measured by radioligand binding techniques. Comp Biochem Physiol C Toxicol Pharmacol. 2004;137:349–353. doi: 10.1016/j.cca.2004.03.002. [DOI] [PubMed] [Google Scholar]
  • 71.Vermoesen K, Serruys AS, Loyens E, Afrikanova T, Massie A, Schallier A, Michotte Y, Crawford AD, Esguerra CV, de Witte PA, Smolders I, Clinckers R. Assessment of the convulsant liability of antidepressants using zebrafish and mouse seizure models. Epilepsy Behav. 2011;22:450–460. doi: 10.1016/j.yebeh.2011.08.016. [DOI] [PubMed] [Google Scholar]
  • 72.Eddins D, Cerutti D, Williams P, Linney E, Levin ED. Zebrafish provide a sensitive model of persisting neurobehavioral effects of developmental chlorpyrifos exposure: comparison with nicotine and pilocarpine effects and relationship to dopamine deficits. Neurotoxicol Teratol. 2010;32:99–108. doi: 10.1016/j.ntt.2009.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kim YH, Lee Y, Lee K, Lee T, Kim YJ, Lee CJ. Reduced neuronal proliferation by proconvulsant drugs in the developing zebrafish brain. Neurotoxicol Teratol. 2010;32:551–557. doi: 10.1016/j.ntt.2010.04.054. [DOI] [PubMed] [Google Scholar]
  • 74.Parsons CG, Stoffler A, Danysz W. Memantine: a NMDA receptor antagonist that improves memory by restoration of homeostasis in the glutamatergic system--too little activation is bad, too much is even worse. Neuropharmacology. 2007;53:699–723. doi: 10.1016/j.neuropharm.2007.07.013. [DOI] [PubMed] [Google Scholar]
  • 75.Greenamyre JT, Young AB. Excitatory amino acids and Alzheimer's disease. Neurobiol Aging. 1989;10:593–602. doi: 10.1016/0197-4580(89)90143-7. [DOI] [PubMed] [Google Scholar]
  • 76.Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC, Yarasheski KE, Bateman RJ. Decreased clearance of CNS beta-amyloid in Alzheimer's disease. Science. 2010;330:1774. doi: 10.1126/science.1197623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Swain HA, Sigstad C, Scalzo FM. Effects of dizocilpine (MK-801) on circling behavior, swimming activity, and place preference in zebrafish (Danio rerio) Neurotoxicol Teratol. 2004;26:725–729. doi: 10.1016/j.ntt.2004.06.009. [DOI] [PubMed] [Google Scholar]
  • 78.Edwards JG, Michel WC. Odor-stimulated glutamatergic neurotransmission in the zebrafish olfactory bulb. J Comp Neurol. 2002;454:294–309. doi: 10.1002/cne.10445. [DOI] [PubMed] [Google Scholar]
  • 79.Ng MC, Tang TH, Ko MC, Wu YJ, Hsu CP, Yang YL, Lu KT. Stimulation of the lateral division of the dorsal telencephalon induces synaptic plasticity in the medial division of adult zebrafish. Neurosci Lett. 2012;512:109–113. doi: 10.1016/j.neulet.2012.01.070. [DOI] [PubMed] [Google Scholar]
  • 80.Alfaro JM, Ripoll-Gomez J, Burgos JS. Kainate administered to adult zebrafish causes seizures similar to those in rodent models. Eur J Neurosci. 2011;33:1252–1255. doi: 10.1111/j.1460-9568.2011.07622.x. [DOI] [PubMed] [Google Scholar]
  • 81.Tiedeken JA, Ramsdell JS, Ramsdell AF. Developmental toxicity of domoic acid in zebrafish (Danio rerio) Neurotoxicol Teratol. 2005;27:711–717. doi: 10.1016/j.ntt.2005.06.013. [DOI] [PubMed] [Google Scholar]
  • 82.Peng YG, Taylor TB, Finch RE, Switzer RC, Ramsdell JS. Neuroexcitatory and neurotoxic actions of the amnesic shellfish poison, domoic acid. Neuroreport. 1994;5:981–985. doi: 10.1097/00001756-199404000-00032. [DOI] [PubMed] [Google Scholar]
  • 83.Tiedeken JA, Ramsdell JS. Embryonic exposure to domoic Acid increases the susceptibility of zebrafish larvae to the chemical convulsant pentylenetetrazole. Environ Health Perspect. 2007;115:1547–1552. doi: 10.1289/ehp.10344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Best JD, Berghmans S, Hunt JJ, Clarke SC, Fleming A, Goldsmith P, Roach AG. Non-associative learning in larval zebrafish. Neuropsychopharmacology. 2008;33:1206–1215. doi: 10.1038/sj.npp.1301489. [DOI] [PubMed] [Google Scholar]
  • 85.Chen J, Patel R, Friedman TC, Jones KS. The Behavioral and Pharmacological Actions of NMDA Receptor Antagonism are Conserved in Zebrafish Larvae. Int J Comp Psychol. 2010;23:82–90. [PMC free article] [PubMed] [Google Scholar]
  • 86.Brown TC, Cole WH, Murray GH. Ketamine: A new anaesthetic agent. Aust N Z J Surg. 1970;39:305–310. [PubMed] [Google Scholar]
  • 87.Finck AD, Ngai SH. Opiate receptor mediation of ketamine analgesia. Anesthesiology. 1982;56:291–297. doi: 10.1097/00000542-198204000-00011. [DOI] [PubMed] [Google Scholar]
  • 88.Zakhary SM, Ayubcha D, Ansari F, Kamran K, Karim M, Leheste JR, Horowitz JM, Torres G. A behavioral and molecular analysis of ketamine in zebrafish. Synapse. 2011;65:160–167. doi: 10.1002/syn.20830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Riehl R, Kyzar E, Allain A, Green J, Hook M, Monnig L, Rhymes K, Roth A, Pham M, Razavi R, Dileo J, Gaikwad S, Hart P, Kalueff AV. Behavioral and physiological effects of acute ketamine exposure in adult zebrafish. Neurotoxicol Teratol. 2011;33:658–667. doi: 10.1016/j.ntt.2011.05.011. [DOI] [PubMed] [Google Scholar]
  • 90.Kanungo J, Cuevas E, Ali SF, Paule MG. Ketamine induces motor neuron toxicity and alters neurogenic and proneural gene expression in zebrafish. J Appl Toxicol. 2011 doi: 10.1002/jat.1751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Stehr CM, Linbo TL, Incardona JP, Scholz NL. The developmental neurotoxicity of fipronil: notochord degeneration and locomotor defects in zebrafish embryos and larvae. Toxicol Sci. 2006;92:270–278. doi: 10.1093/toxsci/kfj185. [DOI] [PubMed] [Google Scholar]
  • 92.Leweke FM, Louvel J, Rausche G, Heinemann U. Effects of pentetrazol on neuronal activity and on extracellular calcium concentration in rat hippocampal slices. Epilepsy Res. 1990;6:187–198. doi: 10.1016/0920-1211(90)90072-4. [DOI] [PubMed] [Google Scholar]
  • 93.Baraban SC, Taylor MR, Castro PA, Baier H. Pentylenetetrazole induced changes in zebrafish behavior, neural activity and c-fos expression. Neuroscience. 2005;131:759–768. doi: 10.1016/j.neuroscience.2004.11.031. [DOI] [PubMed] [Google Scholar]
  • 94.Pearl PL, Shukla L, Theodore WH, Jakobs C, Michael Gibson K. Epilepsy in succinic semialdehyde dehydrogenase deficiency, a disorder of GABA metabolism. Brain Dev. 2011;33:796–805. doi: 10.1016/j.braindev.2011.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wong K, Stewart A, Gilder T, Wu N, Frank K, Gaikwad S, Suciu C, Dileo J, Utterback E, Chang K, Grossman L, Cachat J, Kalueff AV. Modeling seizure-related behavioral and endocrine phenotypes in adult zebrafish. Brain Res. 2010;1348:209–215. doi: 10.1016/j.brainres.2010.06.012. [DOI] [PubMed] [Google Scholar]
  • 96.Lee Y, Kim D, Kim YH, Lee H, Lee CJ. Improvement of pentylenetetrazol-induced learning deficits by valproic acid in the adult zebrafish. Eur J Pharmacol. 2010;643:225–231. doi: 10.1016/j.ejphar.2010.06.041. [DOI] [PubMed] [Google Scholar]
  • 97.Ellis LD, Seibert J, Soanes KH. Distinct models of induced hyperactivity in zebrafish larvae. Brain Res. 2012 doi: 10.1016/j.brainres.2012.02.022. [DOI] [PubMed] [Google Scholar]
  • 98.Donnini S, Solito R, Cetti E, Corti F, Giachetti A, Carra S, Beltrame M, Cotelli F, Ziche M. Abeta peptides accelerate the senescence of endothelial cells in vitro and in vivo, impairing angiogenesis. Faseb J. 2010;24:2385–2395. doi: 10.1096/fj.09-146456. [DOI] [PubMed] [Google Scholar]
  • 99.Chambon C, Wegener N, Gravius A, Danysz W. Behavioural and cellular effects of exogenous amyloid-beta peptides in rodents. Behav Brain Res. 2011;225:623–641. doi: 10.1016/j.bbr.2011.08.024. [DOI] [PubMed] [Google Scholar]
  • 100.Braidy N, Munoz P, Palacios AG, Castellano-Gonzalez G, Inestrosa NC, Chung RS, Sachdev P, Guillemin GJ. Recent rodent models for Alzheimer's disease: clinical implications and basic research. J Neural Transm. 2012;119:173–195. doi: 10.1007/s00702-011-0731-5. [DOI] [PubMed] [Google Scholar]
  • 101.Obulesu M, Venu R, Somashekhar R. Tau mediated neurodegeneration: an insight into Alzheimer's disease pathology. Neurochem Res. 2011;36:1329–1335. doi: 10.1007/s11064-011-0475-5. [DOI] [PubMed] [Google Scholar]
  • 102.Sierra S, Ramos MC, Molina P, Esteo C, Vazquez JA, Burgos JS. Statins as neuroprotectants: a comparative in vitro study of lipophilicity, blood-brain-barrier penetration, lowering of brain cholesterol, and decrease of neuron cell death. J Alzheimers Dis. 2011;23:307–318. doi: 10.3233/JAD-2010-101179. [DOI] [PubMed] [Google Scholar]
  • 103.Zhang Z, Simpkins JW. An okadaic acid-induced model of tauopathy and cognitive deficiency. Brain Res. 2010;1359:233–246. doi: 10.1016/j.brainres.2010.08.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Costa AP, Tramontina AC, Biasibetti R, Batassini C, Lopes MW, Wartchow KM, Bernardi C, Tortorelli LS, Leal RB, Goncalves CA. Neuroglial alterations in rats submitted to the okadaic acid-induced model of dementia. Behav Brain Res. 2012;226:420–427. doi: 10.1016/j.bbr.2011.09.035. [DOI] [PubMed] [Google Scholar]
  • 105.Postlethwait JH, Woods IG, Ngo-Hazelett P, Yan YL, Kelly PD, Chu F, Huang H, Hill-Force A, Talbot WS. Zebrafish comparative genomics and the origins of vertebrate chromosomes. Genome Res. 2000;10:1890–1902. doi: 10.1101/gr.164800. [DOI] [PubMed] [Google Scholar]
  • 106.Sprague J, Bayraktaroglu L, Bradford Y, Conlin T, Dunn N, Fashena D, Frazer K, Haendel M, Howe DG, Knight J, Mani P, Moxon SA, Pich C, Ramachandran S, Schaper K, Segerdell E, Shao X, Singer A, Song P, Sprunger B, Van Slyke CE, Westerfield M. The Zebrafish Information Network: the zebrafish model organism database provides expanded support for genotypes and phenotypes. Nucleic Acids Res. 2008;36:D768–772. doi: 10.1093/nar/gkm956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kyzar E, Zapolsky I, Green J, Gaikwad S, Pham M, Collins C, Roth A, Stewart AM, St-Pierre P, Hirons B, Kalueff AV. The Zebrafish Neurophenome Database (ZND): A Dynamic Open-Access Resource for Zebrafish Neurophenotypic Data. Zebrafish. 2011;9:8–14. doi: 10.1089/zeb.2011.0725. [DOI] [PubMed] [Google Scholar]
  • 108.Selkoe DJ. The molecular pathology of Alzheimer's disease. Neuron. 1991;6:487–498. doi: 10.1016/0896-6273(91)90052-2. [DOI] [PubMed] [Google Scholar]
  • 109.Haass C. Take five--BACE and the gamma-secretase quartet conduct Alzheimer's amyloid beta-peptide generation. Embo J. 2004;23:483–488. doi: 10.1038/sj.emboj.7600061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Iwatsubo T, Odaka A, Suzuki N, Mizusawa H, Nukina N, Ihara Y. Visualization of A beta 42(43) and A beta 40 in senile plaques with end-specific A beta monoclonals: evidence that an initially deposited species is A beta 42(43) Neuron. 1994;13:45–53. doi: 10.1016/0896-6273(94)90458-8. [DOI] [PubMed] [Google Scholar]
  • 111.Musa A, Lehrach H, Russo VA. Distinct expression patterns of two zebrafish homologues of the human APP gene during embryonic development. Dev Genes Evol. 2001;211:563–567. doi: 10.1007/s00427-001-0189-9. [DOI] [PubMed] [Google Scholar]
  • 112.Guo Q, Wang Z, Li H, Wiese M, Zheng H. APP physiological and pathophysiological functions: insights from animal models. Cell Res. 2011;22:78–89. doi: 10.1038/cr.2011.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lee JA, Cole GJ. Generation of transgenic zebrafish expressing green fluorescent protein under control of zebrafish amyloid precursor protein gene regulatory elements. Zebrafish. 2007;4:277–286. doi: 10.1089/zeb.2007.0516. [DOI] [PubMed] [Google Scholar]
  • 114.Joshi P, Liang JO, DiMonte K, Sullivan J, Pimplikar SW. Amyloid precursor protein is required for convergent-extension movements during Zebrafish development. Dev Biol. 2009;335:1–11. doi: 10.1016/j.ydbio.2009.07.041. [DOI] [PubMed] [Google Scholar]
  • 115.Capell A, Steiner H, Romig H, Keck S, Baader M, Grim MG, Baumeister R, Haass C. Presenilin-1 differentially facilitates endoproteolysis of the beta-amyloid precursor protein and Notch. Nat Cell Biol. 2000;2:205–211. doi: 10.1038/35008626. [DOI] [PubMed] [Google Scholar]
  • 116.Selkoe DJ, Podlisny MB. Deciphering the genetic basis of Alzheimer's disease. Annu Rev Genomics Hum Genet. 2002;3:67–99. doi: 10.1146/annurev.genom.3.022502.103022. [DOI] [PubMed] [Google Scholar]
  • 117.Walker ES, Martinez M, Brunkan AL, Goate A. Presenilin 2 familial Alzheimer's disease mutations result in partial loss of function and dramatic changes in Abeta 42/40 ratios. J Neurochem. 2005;92:294–301. doi: 10.1111/j.1471-4159.2004.02858.x. [DOI] [PubMed] [Google Scholar]
  • 118.Leimer U, Lun K, Romig H, Walter J, Grunberg J, Brand M, Haass C. Zebrafish (Danio rerio) presenilin promotes aberrant amyloid beta-peptide production and requires a critical aspartate residue for its function in amyloidogenesis. Biochemistry. 1999;38:13602–13609. doi: 10.1021/bi991453n. [DOI] [PubMed] [Google Scholar]
  • 119.Groth C, Nornes S, McCarty R, Tamme R, Lardelli M. Identification of a second presenilin gene in zebrafish with similarity to the human Alzheimer's disease gene presenilin2. Dev Genes Evol. 2002;212:486–490. doi: 10.1007/s00427-002-0269-5. [DOI] [PubMed] [Google Scholar]
  • 120.Nornes S, Groth C, Camp E, Ey P, Lardelli M. Developmental control of Presenilin1 expression, endoproteolysis, and interaction in zebrafish embryos. Exp Cell Res. 2003;289:124–132. doi: 10.1016/s0014-4827(03)00257-x. [DOI] [PubMed] [Google Scholar]
  • 121.Campbell WA, Yang H, Zetterberg H, Baulac S, Sears JA, Liu T, Wong ST, Zhong TP, Xia W. Zebrafish lacking Alzheimer presenilin enhancer 2 (Pen-2) demonstrate excessive p53-dependent apoptosis and neuronal loss. J Neurochem. 2006;96:1423–1440. doi: 10.1111/j.1471-4159.2006.03648.x. [DOI] [PubMed] [Google Scholar]
  • 122.van Tijn P, Kamphuis W, Marlatt MW, Hol EM, Lucassen PJ. Presenilin mouse and zebrafish models for dementia: focus on neurogenesis. Prog Neurobiol. 2011;93:149–164. doi: 10.1016/j.pneurobio.2010.10.008. [DOI] [PubMed] [Google Scholar]
  • 123.Nornes S, Newman M, Verdile G, Wells S, Stoick-Cooper CL, Tucker B, Frederich-Sleptsova I, Martins R, Lardelli M. Interference with splicing of Presenilin transcripts has potent dominant negative effects on Presenilin activity. Hum Mol Genet. 2008;17:402–412. doi: 10.1093/hmg/ddm317. [DOI] [PubMed] [Google Scholar]
  • 124.Nornes S, Newman M, Wells S, Verdile G, Martins RN, Lardelli M. Independent and cooperative action of Psen2 with Psen1 in zebrafish embryos. Exp Cell Res. 2009;315:2791–2801. doi: 10.1016/j.yexcr.2009.06.023. [DOI] [PubMed] [Google Scholar]
  • 125.De Strooper B. Aph-1, Pen-2, and Nicastrin with Presenilin generate an active gamma-Secretase complex. Neuron. 2003;38:9–12. doi: 10.1016/s0896-6273(03)00205-8. [DOI] [PubMed] [Google Scholar]
  • 126.Edbauer D, Winkler E, Regula JT, Pesold B, Steiner H, Haass C. Reconstitution of gamma-secretase activity. Nat Cell Biol. 2003;5:486–488. doi: 10.1038/ncb960. [DOI] [PubMed] [Google Scholar]
  • 127.De Strooper B, Annaert W. Novel research horizons for presenilins and gamma-secretases in cell biology and disease. Annu Rev Cell Dev Biol. 2010;26:235–260. doi: 10.1146/annurev-cellbio-100109-104117. [DOI] [PubMed] [Google Scholar]
  • 128.Sager JJ, Bai Q, Burton EA. Transgenic zebrafish models of neurodegenerative diseases. Brain Struct Funct. 2010;214:285–302. doi: 10.1007/s00429-009-0237-1. [DOI] [PubMed] [Google Scholar]
  • 129.Francis R, McGrath G, Zhang J, Ruddy DA, Sym M, Apfeld J, Nicoll M, Maxwell M, Hai B, Ellis MC, Parks AL, Xu W, Li J, Gurney M, Myers RL, Himes CS, Hiebsch R, Ruble C, Nye JS, Curtis D. aph-1 and pen-2 are required for Notch pathway signaling, gamma-secretase cleavage of betaAPP, and presenilin protein accumulation. Dev Cell. 2002;3:85–97. doi: 10.1016/s1534-5807(02)00189-2. [DOI] [PubMed] [Google Scholar]
  • 130.Avila J, Lucas JJ, Perez M, Hernandez F. Role of tau protein in both physiological and pathological conditions. Physiol Rev. 2004;84:361–384. doi: 10.1152/physrev.00024.2003. [DOI] [PubMed] [Google Scholar]
  • 131.Brandt R, Hundelt M, Shahani N. Tau alteration and neuronal degeneration in tauopathies: mechanisms and models. Biochim Biophys Acta. 2005;1739:331–354. doi: 10.1016/j.bbadis.2004.06.018. [DOI] [PubMed] [Google Scholar]
  • 132.Wang JZ, Liu F. Microtubule-associated protein tau in development, degeneration and protection of neurons. Prog Neurobiol. 2008;85:148–175. doi: 10.1016/j.pneurobio.2008.03.002. [DOI] [PubMed] [Google Scholar]
  • 133.Chun W, Johnson GV. The role of tau phosphorylation and cleavage in neuronal cell death. Front Biosci. 2007;12:733–756. doi: 10.2741/2097. [DOI] [PubMed] [Google Scholar]
  • 134.Bai Q, Burton EA. Zebrafish models of Tauopathy. Biochim Biophys Acta. 2011;1812:353–363. doi: 10.1016/j.bbadis.2010.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Chen M, Martins RN, Lardelli M. Complex splicing and neural expression of duplicated tau genes in zebrafish embryos. J Alzheimers Dis. 2009;18:305–317. doi: 10.3233/JAD-2009-1145. [DOI] [PubMed] [Google Scholar]
  • 136.Bai Q, Garver JA, Hukriede NA, Burton EA. Generation of a transgenic zebrafish model of Tauopathy using a novel promoter element derived from the zebrafish eno2 gene. Nucleic Acids Res. 2007;35:6501–6516. doi: 10.1093/nar/gkm608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Tomasiewicz HG, Flaherty DB, Soria JP, Wood JG. Transgenic zebrafish model of neurodegeneration. J Neurosci Res. 2002;70:734–745. doi: 10.1002/jnr.10451. [DOI] [PubMed] [Google Scholar]
  • 138.Paquet D, Bhat R, Sydow A, Mandelkow EM, Berg S, Hellberg S, Falting J, Distel M, Koster RW, Schmid B, Haass C. A zebrafish model of tauopathy allows in vivo imaging of neuronal cell death and drug evaluation. J Clin Invest. 2009;119:1382–1395. doi: 10.1172/JCI37537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Saunders AM, Strittmatter WJ, Schmechel D, George-Hyslop PH, Pericak-Vance MA, Joo SH, Rosi BL, Gusella JF, Crapper-MacLachlan DR, Alberts MJ, et al. Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer's disease. Neurology. 1993;43:1467–1472. doi: 10.1212/wnl.43.8.1467. [DOI] [PubMed] [Google Scholar]
  • 140.Verghese PB, Castellano JM, Holtzman DM. Apolipoprotein E in Alzheimer's disease and other neurological disorders. Lancet Neurol. 2011;10:241–252. doi: 10.1016/S1474-4422(10)70325-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kim J, Basak JM, Holtzman DM. The role of apolipoprotein E in Alzheimer's disease. Neuron. 2009;63:287–303. doi: 10.1016/j.neuron.2009.06.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Durliat M, Andre M, Babin PJ. Conserved protein motifs and structural organization of a fish gene homologous to mammalian apolipoprotein E. Eur J Biochem. 2000;267:549–559. doi: 10.1046/j.1432-1327.2000.01033.x. [DOI] [PubMed] [Google Scholar]
  • 143.Babin PJ, Thisse C, Durliat M, Andre M, Akimenko MA, Thisse B. Both apolipoprotein E and A-I genes are present in a nonmammalian vertebrate and are highly expressed during embryonic development. Proc Natl Acad Sci U S A. 1997;94:8622–8627. doi: 10.1073/pnas.94.16.8622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Monnot MJ, Babin PJ, Poleo G, Andre M, Laforest L, Ballagny C, Akimenko MA. Epidermal expression of apolipoprotein E gene during fin and scale development and fin regeneration in zebrafish. Dev Dyn. 1999;214:207–215. doi: 10.1002/(SICI)1097-0177(199903)214:3<207::AID-AJA4>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Neurodegenerative Disease are provided here courtesy of e-Century Publishing Corporation

RESOURCES