1-s2.0-S0161813X1300079X-main in

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

NeuroToxicology 37 (2013) 163–172

Contents lists available at SciVerse ScienceDirect

NeuroToxicology

Review

Okadaic acid induced neurotoxicity: An emerging tool to study Alzheimer’s


disease pathology
Pradip K. Kamat a,*, Shivika Rai b, Chandishwar Nath b
a
Division of Physiology and Biophysics, University of Louisville, School of Medicine, KY 40202, United States
b
Division of Pharmacology Central Drug Research Institute (CDRI), P.O. Box 173, Lucknow, U.P. 226001, India

A R T I C L E I N F O A B S T R A C T

Article history: Okadaic acid (OKA) is one of the main polyether toxins produced by marine microalgae which causes
Received 1 January 2013 diarrhetic shellfish poisoning. It is a selective and potent inhibitor of serine/threonine phosphatases 1
Accepted 3 May 2013 and 2A induces hyperphosphorylation of tau in vitro and in vivo. The reduced activity of phosphatases
Available online 17 May 2013
like, protein phosphatase 2A (PP2A) has been implicated in the brain of Alzheimer’s disease (AD)
patients. It is reported that AD is a complex multifactorial neurodegenerative disorder and
Keywords: hyperphosphorylated tau proteins is a major pathological hallmark of AD. The molecular pathogenesis
Okadaic acid
of AD includes an extracellular deposition of beta amyloid (Ab), accumulation of intracellular
Alzheimer’s disease
Neurotoxicity
neurofibrillary tangles (NFT), GSK3b activation, oxidative stress, altered neurotransmitter and
inflammatory cascades. Several lines of evidence suggested that the microinfusion of OKA into the
rat brain causes cognitive deficiency, NFTs-like pathological changes and oxidative stress as seen in AD
pathology via tau hyperphosphorylation caused by inhibition of protein phosphatases. So, communal
data and information inferred that OKA induces neurodegeneration along with tau hyperpho-
sphorylation; GSK3b activation, oxidative stress, neuroinflammation and neurotoxicity which is a
characteristic feature of AD pathology. Through this collected evidence, it is suggested that OKA induced
neurotoxicity may be a novel tool to study Alzheimer’s disease pathology and helpful in development of
new therapeutic approach.
ß 2013 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164
2. Factors responsible for AD pathology and pathological hallmarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164
2.1. Okadaic acid and Alzheimer’s disease pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164
2.2. Tau phosphorylation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165
2.3. Glycogen synthase kinase-3 beta (GSK3b) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
2.4. Kinases involved in Tau pathology. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
3. Physiopathological conditions and factors involved in AD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
3.1. Cholinergic function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
3.2. NMDA receptor function and excitotoxicity: . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168
3.3. Role of mitochondria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168
3.4. Oxidative stress. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168
3.5. Neuroinflammation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169
3.6. OKA induced neurotoxicity and cell death . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169
4. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169
5. Future prospectus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170

Abbreviations: OKA, Okadaic acid; PP2A, Protein phosphatase 2A; GSK-3b, Glycogen synthase kinase-3b; PHF, Paired helical filament; NFT, Neurofibrillary tangle; AD,
Alzheimer’s disease; Ab, Beta amyloid; AChE, Acetylcholinestrse activity; ChAT, Choline acetyle transferase activity; ACh, Acetylcholine; NMDA, N-methyl D-aspartate; PKA,
Protein kinase; M1,M2,M3, Muscarinic receptors; nAChR, Nicotinic receptor; ERK, Extracellular receptor kinase.
* Corresponding author at: Division of Physiology and Biophysics, University of Louisville, Louisville, KY 40202, United States. Tel.: +1 52 852 3638.
E-mail address: pradeepkamat2004@gmail.com (P.K. Kamat).

0161-813X/$ – see front matter ß 2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.neuro.2013.05.002
164 P.K. Kamat et al. / NeuroToxicology 37 (2013) 163–172

1. Introduction contribute to different stages of the enzymatic cascade believed


to underlie memory formation (Bennett et al., 2001). He et al.
Okadaic acid (OKA) is one of the major substances among (2001) investigated the effects of OKA, on spatial memory and
Diarrheic shellfish poisoning (DSP) toxins worldwide. It was first neuronal survival in rats. It seems that OKA is an extremely useful
isolated from the black sponge Holichondria okadai (Takai et al., tool for studying the cellular processes that are regulated by
1987) and later in the dinoflagellate Prorocentrum lima (Murakami reversible phosphorylation of proteins as signal transduction, cell
et al., 1994) and in Dinophysis acuta (Garcia et al., 2004). OKA is division and memory (Fernandez et al., 2002).
known to be a potent inhibitor of Ser/Thr phosphatases and a In this review, we discussed that why OKA induced neurotox-
specific inhibitor of protein synthesis by hyperphosphorylation of icity and molecular alteration is better tool to study AD pathology.
elongation factor EF2 (Bialojan and Takai, 1988). Previous studies There are several factors responsible for AD disease pathology but
have shown that OKA is widely distributed in mammals after tau-hyper phosphorylation is a main pathological hallmark.
consumption (Matias and Creppy, 1996b), the main effects being in Several reports reveal that same pathology is impersonated by
the intestinal tract where it is responsible for DSP (Sueoka et al., OKA in vivo and in vitro study.
1997). There is clinical observation that OKA causes memory
impairment in human subjects those who used seafood contami- 2. Factors responsible for AD pathology and pathological
nated with dinoflagellate (Helicondria okadai) (http://www.aris- hallmarks
tatek.com/Newsletter/DEC07/DEC07ts.aspx). Due to its inhibitory
action on protein phosphatases 1 and 2A, OKA used as a tool for 2.1. Okadaic acid and Alzheimer’s disease pathology
studying the participation of protein phosphorylation in cellular
processes (Arias et al., 1998). Ser/Thr protein phosphatases, PP1 AD is a progressive neurodegenerative disease that causes
and PP2A, can dephosphorylate tau in vitro. In AD brain, the cognitive and behavioral deterioration in the aged person. In AD
activity of PP2A appears to be reduced in both gray and white brain, the activity of PP2A appears to be reduced in both gray and
matters (Gong et al., 1993; Torrent and Ferrer, 2012) and a down- white matters (Gong et al., 1993) and a down-regulation of this
regulation of this enzyme are apparently involved in slowing down enzyme are apparently involved in slowing down the process of
the process of tau dephosphorylation (Matsuo et al., 1994). tau dephosphorylation (Matsuo et al., 1994). AD is a common
Alzheimer’s disease (AD) is the most common form of dementia neurodegenerative disease pathologically by histopathological
and characterized by severe neurodegenerative changes, such as changes including extracellular deposits of amyloid-beta (Ab)
cerebral atrophy, loss of neurons and synapses, and selective peptides forming senile plaques (SP) and the intracellular
depletion of neurotransmitter systems in cerebral cortex and neurofibrillary tangles (NFT) of hyperphosphorylated tau in the
certain subcortical region. Brain regions associated with higher brain. Natalia et al. (2012) reported that the hyperphosphorylation
mental functions, particularly the neocortex and hippocampus are of tau protein and formation of intraneuronal neurofibrillary
most affected in AD pathology (Hampel et al., 2008). AD pathology tangles (NFTs) represents another neuropathological hallmark in
includes the extracellular deposits of b-amyloid (derived from AD brain. Tau is a microtubule-associated protein and localizes
amyloid precursor protein; APP) in senile plaques (Nunomura predominantly in the axons of neurons with the primary function
et al., 2010), intracellular formation of NFT (containing an in maintaining microtubules stability. Neurofibrillary tangles
abnormally phosphorylated form of a microtubule associated (NFTs) are one of the pathological hallmarks of AD that has been
protein, tau) (Binder et al., 2005), and the loss of neuronal synapses shown to correlate positively with the severity of dementia in the
and pyramidal neurons (Capetillo-Zarate et al., 2012). AD is neocortex of AD patients. Chopra et al. (2011) also reported that Ab
associated with neurodegenerative changes which compromise containing senile plaques and phospho-tau-containing neurofi-
not only cognitive functioning but also lead to a decline in brillary tangles are hallmark lesions of AD pathology. OKA, when
functional abilities and induce a spectrum of psychological or injected into the right lateral dorsal hippocampus area of adult rat
behavioral symptoms (Radebaugh et al., 1996). Many efforts are brain depicts an in vivo model of AD tauopathy (Zhang et al., 2010a,
currently undertaken to investigate AD pathology and develop b). Zhou et al. (2008) suggested that the increased demethylation
appropriate treatment strategies. These strategies focus on long- of PP2A mediated by Ab overproduction may contribute to the
term preservation of cognitive and functional abilities or slowing reduced PP2A activity resulting in the compromised dephosphor-
down disease progression along with reducing behavioral symp- ylation of abnormally hyperphosphorylated tau. Hyperphosphory-
toms and maintaining the patient’s quality of life (Geldmacher, lated tau, which is the major protein of the neurofibrillary tangles
2007). These changes result in the development of the typical in AD brain, is most probably the result of an imbalance of tau
symptomology of AD characterized by gross and progressive kinase and phosphatase activities in the affected neurons (Gong
impairments of cognitive function and often accompanied by et al., 2000). Selective inhibition of protein phosphatase 2A by OKA
behavioral disturbances such as aggression and depression. AD induced an AD-like hyperphosphorylation and accumulation of tau
together with other forms of dementia represents a major (Gong et al., 2000). The hyperphosphorylated tau had a reduced
challenge for health care systems with aging populations. ability to bind to microtubules and to promote microtubule
Memory impairment induced by intra-hippocampal injection of assembly in vitro (Ballatore et al., 2012). The biochemical evidence
OKA has been reported, accompanied by remarkable neuropatho- unambiguously demonstrates the hyperphosphorylation and the
logical changes including hippocampal neurodegeneration, a accumulation of neurofibril (NF) subunits in AD brain (Fig. 1). The
paired helical filament-like phosphorylation of tau protein, and inhibition of PP-2A/PP-1 activities can lead to the hyperpho-
formation of b-amyloid containing plaque-like structures (Costa sphorylation of NF (Wang et al., 2001).
et al., 2012). Wu et al. (2008) found that the injection of OKA into Zhao et al. (1994) and subsequently; our group (Kamat et al.,
rat lateral ventricles constitutes a promising animal model for 2010) also reported that OKA (ICV) causes spatial memory
investigating selective aspects of AD. Maidana et al. (2006) impairment in rat. Microtubule-associated protein tau is abnor-
investigated OKA affects short- and long-term memory alteration mally hyperphosphorylated, glycosylated, and aggregated in
in rats and caused oxidative damage in hippocampus of rats. Acute affected neurons in AD and found that the aberrant tau
administration of various concentrations of OKA was found to glycosylation precedes tau hyperphosphorylation in AD brain
disrupt two distinct stages of memory formation in day old chick (Liu and Wang, 2002). The phosphorylation state of proteins
(Bennett et al., 2003). Different phosphatase enzymes may modulated by protein kinases and protein phosphatases is
P.K. Kamat et al. / NeuroToxicology 37 (2013) 163–172 165

Fig. 1. Schematic diagram showing the involvement of OKA in the pathogenesis of Alzheimer’s disease. OKA induce APP mismetabolism causing an increase in Ab which
results in neurite plaque formation, microglial activation, oxidative stress and neuroinflammation. OKA induced Ab may also precede formation of neurofibrillary tangles.
Neuritic plaques and neurofibrillary tangles lead to neurotoxicity, neuronal dysfunction and finally to AD like pathology.

important in the regulation of many physiological processes like brain regions involved in learning memory function (He et al.,
the release of neurotransmitters and activation of postsynaptic 2005). In vitro, OKA treatment in neuroblastoma SKNSH-SY 5Y
receptors (Huganir and Greengard, 1990). Tau phosphorylation cells also produced tau proteins with an increased apparent
also involve in maintenance of long-term potentiation (Bliss and molecular weight and a more acidic isoelectric point when
Collingridge, 1993), and learning and memory (Zhao et al., 1994). compared to Tau proteins from control cells (Suuronen et al.,
Immunocytochemical staining revealed hyperphosphorylated tau 2000). OKA also induces neurotoxicity of in SH-SY5Y cell line and
accumulation in pyramidal neurons in cornu ammonis and in cultured neuronal cells which developed similar pathological
neocortical neurons. The topography of these changes recalls the condition like AD (Zhang et al., 2007).
distribution of NFT in AD brain (van de Nes et al., 2006). The
discovery that rare mutations in the gene encoding for APP always 2.2. Tau phosphorylation
led to AD in family members carrying the defect resulted in the
proposal of the ‘‘amyloid cascade hypothesis’’ of AD (Pera et al., Tau proteins are microtubule-associated proteins in neurons
2013). Levine et al. (1995) found that injection of the phosphatase and under physiological condition, tau plays a key role in
inhibitor OKA results in modulation of synaptic transmission in the microtubules stabilization, axonal transportation and neurite
hippocampus via brain-derived neurotrophic factor (BDNF). BDNF outgrowth (Yi and Simpkins, 2008). Tau hyperphosphorylation
which is associated with memory rapidly enhanced synaptic and memory deficit are characteristic alterations of AD. Inhibition
efficacy through increased postsynaptic responsiveness via a of PP2A through hippocampal injection of OKA induces tau
phosphorylation-dependent pathway. It has been reported that hyperphosphorylation and memory impairment in rats (Yin
impairment of APP metabolism leading to increased production of et al., 2010). Hyperphosphorylated proteins have been implicated
Ab was proposed as the critical event in with other changes, in the neurodegenerative disorder AD one of the most character-
tangles, neuron loss, synapse loss, and neurotransmission dys- istics histopathological features of AD is NFT, which contains
function (Wang et al., 2012). These neurons therefore seem to have distorted microtubule and phosphorylated tau proteins (Reynolds
a central role in the clinical symptoms as well as in the et al., 2008). Under pathological conditions, deposits of abnormally
pathophysiology of the AD disease. Arias et al. (1998) found the hyperphosphorylated tau protein are diagnostic hallmark of AD.
hyperphosphorylation of tau protein due to inhibition of phos- Tau, as the principal component of neurofibrillary tangles, can be
phatases in vivo induces neuronal stress and subsequent hyperphosphorylated by a reduced activity of PP2A in vitro and by
neurodegeneration. OKA-induced spatial memory impairment is pharmacological approaches, suggesting a crucial role of PP2A in
associated with neuropathological changes like loss of pyramidal tangle formation in AD (Ballatore et al., 2007). Most of the
cell in the CAI and apoptotic cell death in the hippocampus, the phosphorylation sites on tau are present in the proline-rich and the
166 P.K. Kamat et al. / NeuroToxicology 37 (2013) 163–172

C-terminal regions flanking the microtubule binding domains persistent activation of ERK1/2 and/or protein kinase C and a
which interact with several kinases (Sergeant et al., 2008). The resulting oxidative stress (Yi et al., 2009). Cerebral OKA adminis-
mechanisms operative in the onset and progression of neurode- tration induces AD like phenotype in rats and phosphorylated tau
generative diseases, such as AD likely involve the disruption of as well as cyclin-dependent kinase 5 (cdk5) and its co activator,
normal phosphorylation events as a result in tau dysfunction p25, were significantly increased in these regions of the brain.
(Abraha et al., 2000). Although the precise reason for the Alterations in glutamate levels associated with hyperactivation of
accumulation of tau in AD is not known, evidence indicates that Cdk5 signaling pathway and tau phosphorylation may participate
it may be related to either an increase in kinase activity or a in the genesis of this pathological phenotype (Zimmer et al., 2012).
decrease in phosphatase activity (Dolan and Johnson, 2010). The kinases that phosphorylate tau can be divided into two major
groups, according to motif specificity: proline-directed protein
2.3. Glycogen synthase kinase-3 beta (GSK3b) kinases (PDPK) and non-proline-directed protein kinases (non-
PDPK) (Gendron and Petrucelli, 2009). The PDPK include cyclin-
Glycogen synthase kinase-3 beta (GSK3b) is a pivotal molecule dependent kinase 5(Cdk5), mitogen-activated protein kinase
in the development of AD and involved in the formation of paired (MAPK), and several stress-activated protein kinases (Gendron
helical filament (PHF)-tau. GSK3b is an integral component of the and Petrucelli, 2009). Both Cdk5 and GSK3b co-purify with
NFT deposits that disrupt neuronal function, and a marker of microtubules and phosphorylate tau within a cellular environment
neurodegeneration in AD (Takashima, 2006). The essential (Wagner et al., 1996). The phosphorylation of tau by these kinases
dysfunction appears to be an imbalance in kinase and phosphatase inhibits the ability of tau to promote microtubule assembly and
activities resulting from dysregulation of the phosphorylation/ facilitates the polymerization of tau into PHF (Evans et al., 2000).
dephosphorylation system (Gong et al., 2003). GSK3b induces Among the non-PDPK are cyclic AMP-dependent protein kinase
hyperphosphorylation of tau, NFT formation, neuronal death and (PKA), calcium- and calmodulin-dependent protein kinase II
synaptic loss in the AD brain and induces memory deficits (CaMKII), and microtubule affinity regulating kinase (MARK), the
(Takashima, 2006). Alterations in GSK3b and PP2A have been mammalian homologue of PAR-1 and targets KXGS motifs within
proposed to be involved in the abnormal tau phosphorylation and the microtubule binding repeat domains of tau (Paudel, 1997). OKA
aggregation linked to AD (Martin et al., 2011). GSK3b is recognized caused potassium channel inactivation, which is regulated by
as one of major kinases to phosphorylate tau in AD, thus lots of AD CaMK-II, and may contribute to the electrical memory of the atrial
drug discoveries target GSK3b, however, the inactive form of myocardium (Tessier et al., 2001). Protein phosphatase-mediates
GSK3b which is phosphorylated at serine-9 is increased in AD the regulation of protein kinase C during long-term depression in
brains. Lim et al. (2010) addressed this paradoxical condition of AD the adult hippocampus in vivo. The neural substrates of learning
in rat neurons treated with OKA which inhibits protein PP2A and and memory are thought to involve in long-term changes in
induces tau hyperphosphorylation and cell death. Interestingly, synaptic function, including long-term depression (LTD) of
OKA also induces phosphorylation of GSK3b at serine-9 and other synaptic strength. Thiels et al. (2000) demonstrated that LTD in
substrates including tau, beta-catenin and CRMP2 like in AD brains the adult hippocampus in vivo involves a decrease in PKC activity.
(Lim et al., 2010). Abnormal hyperphosphorylation of tau appears It is mediated, by dephosphorylation of the catalytic domain of PKC
to be crucial in neurofibrillary degeneration in AD (Qian et al., by protein phosphatases activated after LTD inducing stimulation
2010). Inhibition of PP2A with OKA in metabolically active rat which was prevented by OKA. Tau phosphorylation induces its
brain slices caused inhibition of GSK3b via an increase in its dissociation from microtubules and prevents its degradation
phosphorylation (Harris et al., 1993). OKA treatment in human (Dickey et al., 2007). Unbound tau may then be hyperpho-
neuronal and glial cultures severely depleted the content of GSK3b sphorylated by other kinases and in fact, the phosphorylation of
and down regulated the GSK3b activity by Akt-dependent tau by MARK/PAR-1 may be a prerequisite for the action of
inhibition suggest that in primary cultures, Akt selectively downstream kinases, including GSK3b and Cdk5 (Nishimura et al.,
phosphorylates tau at S214 rather than T212 and the possibility 1998) (Fig. 2).
that tau S214 may participate in Akt-mediated anti-apoptotic
signaling. The overall alterations in tau phosphorylation induced 3. Physiopathological conditions and factors involved in AD
by PP2A inhibition were the result of the combined effects of both
reduced tau dephosphorylation due to PP2A inhibition directly and 3.1. Cholinergic function
reduced phosphorylation by GSK3b due to its inhibition. It is
suggest that GSK3b may still have its kinase activity despite AD is characterized by neuronal loss and neurofibrillary tangle
increase of its phosphorylation in AD brains at least in PP2A- formation and primarily affecting pyramidal neurons and their
compromised conditions and that GSK3b inhibitors could be a synapses neurotransmitter specific subcortical nuclei including
valuable drug candidate in AD. the cholinergic nucleus basalis of Meynert and medial septum.
Biochemical investigations of biopsy tissue taken from patients
2.4. Kinases involved in Tau pathology with AD after the onset of symptoms indicate that selective
neurotransmitter pathology occurs early in the course of the
Inactivation of PP1/PP2A via oxidative stress has been shown in disease (Francis, 1996). Specifically, presynaptic markers of the
vitro and in vivo to be involved in hyperphosphorylation of tau and cholinergic system appear uniformly reduced. This is exemplified
prolonged phosphorylation of ERK 1/2 (Rahman et al., 2005; by reductions in Acetylcholinestrse activity (AChE), Choline acetyle
Poppek et al., 2006). Thus, it is intriguing to postulate that transferase (ChAT) activity and Acetylcholine (ACh) synthesis
oxidative stress mediated PP1 and PP2A inhibition in AD may which are strongly correlated with the degree of cognitive
account for enhanced ERK1/2 activity and subsequent tau impairment in patients with AD (Kamat et al., 2012a,b). Moreover,
hyperphosphorylation and neurofibrillary tangle formation. Li a few patients with AD do not show large decreases in ChAT
et al. (2004a,b) suggested that decrease in PP-2A activity could activity, albeit that a small reduction is found in the amygdale
have caused the activation of ERK1/2, MEK1/2, and p70 S6 kinase, (Palmer et al., 1986) Thus, although diminished ChAT activity is a
and the abnormal hyperphosphorylation of tau both via an necessary correlate of AD, additional factors other than impaired
increase in its phosphorylation and a decrease in its dephosphor- cholinergic function are likely to participate in the decline in
ylation in AD brain. OKA induced neurotoxicity are mediated by cognitive function. Other studies have demonstrated a reduction in
P.K. Kamat et al. / NeuroToxicology 37 (2013) 163–172 167

Fig. 2. Schematic diagram depicting the role of kinases and phosphatases acing on Tau protein in OKA induced AD pathology. Tau contains specific phosphorylation sites that
are apparently critical for hyper-phosphorylation of Tau in AD. Proline-directed protein kinases such as MAPKs, GSK3b and Cdk-5 basically phosphorylate Tau protein, and
the non proline-directed protein kinases such as Cdk5 and MAPK. Several other kinases are also phosphorylate the Tau protein such as GSK3b, ERK1/2, MEK1/2, and p70 S6
kinase, CamkII and p25 and finally involved in development of AD pathology.

the number of nicotinic (Whitehouse and Kellar, 1987) and is a characteristic feature of AD and we found that administration
muscarinic (M2) ACh receptors in AD brains, most of which are of OKA in rat brain causes neurodegeneration along with
considered to be located on presynaptic cholinergic terminals, but cholinergic dysfunction (Kamat et al., 2012a,b). It is reported that
a relative preservation of postsynaptic muscarinic (M1, M3 muscarinic cholinergic receptor stimulation and activation of
receptors. It is reported that the injection of OKA into the meynert protein kinase C may lead to the inactivation of a protein kinase
nucleus basalis, cortex and hippocampus of rat brain induces GSK3b, which phosphorylates tau, in vitro, in a similar manner to
decreased acetylcholine level and spatial memory deficit (Tian that found in AD (Lovestone et al., 1994). Neuronal cells culture
et al., 2004). They found that injection of OKA induced hyperpho- transfected with M1 muscarinic receptors show reduced phos-
sphorylation of tau and NF and decreased ACh level in the nucleus phorylation of tau after treatment with cholinergic agonists
basalis of Meynert. These alterations were accompanied by spatial (Sadot et al., 1996). On the basis of the above evidence, neocortical
memory deficit in OKA-injected rats. He et al. (2001) reported that cholinergic innervation is probably lost at an early stage of the AD
OKA into the rat brain causes spatial memory deficit and disease (Dalley et al., 2005). It is reported that both a7 and b2
neurodegeneration in the rat brain. Abnormal hyperphosphoryla- nAChR agonist can afford neuroprotection against OKA induced
tion of tau and cholinergic deficit occur in the early stage of AD and neurotoxicity (Del Barrio et al., 2011). Protection mediated by a7
relate to the dementia symptom (Tian et al., 2004). Further, Tian nAChRs was independent of Ca2+ and involved the intracellular
et al. (2004) also demonstrated that the OKA-induced ACh signaling pathway Janus Kinase-2/Phosphatidylinositol-3-ki-
reduction may be due to a failure of intraneuronal transport of nase/Akt. However, although the loss of cholinergic function is
ChAT from cell body to the neuronal terminals rather than an correlated with the cognitive impairment in AD (Kamat et al.,
alteration of activity of ChAT or acetylcholinesterase. Insulin 2012a,b) an association between two such indices does not
injection into the lateral cerebral ventricle of the rats improve necessarily indicate a causal relation. Cholinergic neurotransmis-
learning abilities in a OKA injected in CA1 region of the rat sion may be a specific target for b-amyloid, as it has been shown to
hippocampus and the expressions of nicotinic acetylcholine reduce both choline uptake and ACh release in vitro (Kar et al.,
receptors were improved (Lin et al., 2012). Cholinergic dysfunction 1998). AD also exhibits extracellular plaques of aggregated
168 P.K. Kamat et al. / NeuroToxicology 37 (2013) 163–172

b-amyloid protein (Ab), intracellular neurofibrillary tangles that 3.3. Role of mitochondria
contain hyperphosphorylated tau protein and a profound loss of
basal forebrain cholinergic neurons that innervate the hippocam- Mitochondria play a critical role in regulation of apoptosis, which
pus and the neocortex (Kar et al., 2004). Pyramidal cells are lost in is implicated in the aging process age-related mitochondrial
the disease, subject to tangle formation, represent a major source oxidative stress may contribute to apoptosis upon aging (Yamaguchi
of APP and are regulated by a neurotransmitter ACh, affected early and Perkins, 2009). Mitochondria are significantly reduced in
in the disease (Francis et al., 1999). Observations in cell lines and various types of cells obtained from patients with AD (Blass et al.,
primary neuronal cultures it is evident that the activation of 1990). Mitochondrial dysfunction is one of the contributory
muscarinic, metabotropic glutamate and other phospholipase C- pathophysiology of AD (Hirai et al., 2001). The most consistent
linked receptors favors the non-amyloidogenic processing of APP defect in mitochondrial electron transport enzymes in AD is a
(Nitsch, 1996). Furthermore, b-amyloid neurotoxicity is attenu- deficiency in cytochrome C oxidase which leads to an increase in ROS
ated by treatment with muscarinic agonists (Emmerling et al., production, a reduction in energy stores and disturbance in energy
1997). No sufficient data are there to report the potential metabolism (Cardoso et al., 2004a,b). OKA induces free radical ROS
beneficial effects of cholinomimetic drugs on either increasing generation, decreased mitochondrial membrane potential and
APP or reducing b-amyloid production in patients with AD. There mitochondrial activity indicates mitochondrial dysfunction in rat
is some report which shows evidence for reductions in CSF fluid brain (Kamat et al., 2011). The activation of the permeability
APP in depressed patients receiving drugs with anticholinergic transition pore in mitochondria, which is believed to play a critical
side effects (Clarke et al., 1993). Therefore, as a consequence of role in cell necrosis and apoptosis, is enhanced in spleen, brain, and
reduced cholinergic activity, reduced protein kinase activity may liver of aged mice (Mather and Rottenberg, 2000). Such a
lead to a higher level of activity of GSK-3 and hence hyperpho- modification of mitochondria may cause the cells more vulnerable
sphorylation of tau. to apoptotic inducers (Zhang and Herman, 2002). Thus, mitochon-
dria appear to influence the aging process via modifying the
3.2. NMDA receptor function and excitotoxicity: regulatory machinery of apoptosis and that mitochondria have a
central role in aging-related neurodegenerative diseases like AD
N-methyl-d-aspartate receptors (NMDARs) are glutamate- (Kamat et al., 2011). Mitochondria are critical regulators of cell
gated ion channels that are highly permeable to Ca2+, and Ca2+ death, a key feature of neurodegeneration. In all major examples of
influx through NMDARs is essential for synaptogenesis, experi- these diseases there is strong evidence that mitochondrial
ence-dependent synaptic remodeling and long-lasting changes in dysfunction occurs early and acts causally in disease pathogenesis
synaptic efficacy such as long-term potentiation and long-term (Kamat et al., 2011). Targeting basic mitochondrial processes, such
depression (Collingridge et al., 2004). The NMDARs is involved in as energy metabolism or free-radical generation, or specific
cellular mechanism for learning and memory function (Rai et al., interactions of disease-related proteins with mitochondria, hold
2012). NMDARs are involved in a wide array of biological great promise for therapies (Kamat et al., 2011). Consistently,
processes which are crucial for brain development and function oxidative stress-induced autophagy of accumulated amyloid b-
(Perez-Otano and Ehlers, 2004). OKA induced dephosphorylation protein in AD causes permeabilization of lysosomal membrane and
of one or more proteins other than adenosine kinase as a leads to neuronal cell death (Zheng et al., 2009). Mitochondria
consequence of NMDA receptor activation might play an damaged by significantly increased oxidative stress in pyramidal
important role in extracellular adenosine regulation, with neurons of AD are subjected to autophagic degradation, ultimately
important consequences for the regulation of excitatory synaptic leading to neurodegeneration (Moreira et al., 2010). An important
transmission, plasticity and excitotoxicity (Lu and Rosenberg, function of mitochondria is to produce ATP and targeting genes
2007). Excitotoxic cell death (ECD) is characteristic of mammalian involved in ATP production offers a great opportunity to study the
brain in several type of neurotoxixity. A key event in ECD is a role of mitochondrial function in aging (Cui et al., 2012).
massive increase in intracellular Ca2+ by over-stimulation of Mitochondrial dysfunction and increased oxidative damage are
NMDARs. Ca2+ and calmodulin play an important role in often associated with neurodegenerative disorder like Alzheimer’s
decreasing NMDAR activity in neurotoxicity (Shin et al., 2005). disease (AD), Parkinson disease (PD) and Huntington’s disease (HD)
Excitotoxicity is defined as a toxic process characterized by a suggesting that oxidative stress may play an important role in the
sustained stimulation of excitatory amino acids receptors pathophysiology of these diseases (Lin and Beal, 2006).
(Nicholls et al., 2007), mainly involving NMDAR. Different toxic
events derived from excitotoxicity have been characterized in 3.4. Oxidative stress
experimental models, including upregulation of detrimental
signaling pathways, disrupted Ca2+ homeostasis, and recruitment Oxidative stress is one of the most important mechanisms
of reactive oxygen/nitrogen species (ROS/RNS), with further involved in toxic events observed in neuronal cells in different
oxidative/nitrosative stress (Sas et al., 2007), ultimately leading to neurodegenerative disorders. Besides the clinical study Cohen
cell death. OKA treatment showed elevated Mitochondrial Ca2+ in (1990) reported in rat brain that OKA selectively inhibits the
rat suggesting OKA might be acting through NMDA system (Kamat serine/threonine phosphatases 1 (PP1) and 2A (PP2A). Arias et al.
et al., 2011, 2013). Immunohistochemical observation suggest (1998) have reported that the protein phosphatase inhibitor OKA
that OKA induces tau phosphorylation in brain slices to prepare induces heat shock protein expression, neurodegeneration in rat
AD models and causes expressions of P-tau, NR1 and NR2B in brain hippocampus in vivo and also found that protein hyperpho-
slices (Li et al., 2004a,b). NMDA-evoked adenosine accumulation sphorylation due to inhibition of phosphatases in vivo induces
is mediated by activation of phosphatase 1/2A and associated with neuronal stress and subsequent neurodegeneration. Oxidative
adenosine kinase inhibition. OKA reduces basal synaptic trans- stress was increased with OKA treatment as measured by Free
mission and blocked the induction of synaptic plasticity in the radical generation and lipid peroxidation (Kamat et al., 2010).
form of long-term potentiation (LTP). LTP involved in memory Several data suggest that the bilateral microinfusion of OKA into
formation and found prolonged inhibition of protein phospha- the rat brain causes cognitive deficiency, NFTs-like pathological
tases and suggests reduced post-synaptic signaling as a major changes, and oxidative stress as seen in AD pathology via tau
mechanism for basal synaptic transmission and LTP impairments hyperphosphorylation caused by inhibition of protein phospha-
(Koss et al., 2007). tases (Kamat et al., 2013). AD includes a variety of risk factors,
P.K. Kamat et al. / NeuroToxicology 37 (2013) 163–172 169

extracellular deposition of b-amyloid, accumulation of intracellu- cytoskeleton that ultimately leads to cell death in primary cortical
lar neurofibrillary tangles, oxidative neuronal damage and neurons and in neuroblastoma cell lines (Arias et al., 1993). In
inflammatory cascades (Chopra et al., 2011). OKA induces cerebellar granule cells, OKA induces disintegration of neurites and
oxidative DNA damage directly and indirectly in SHSY5Y cells, swelling of cell bodies (Fernandez et al., 1991). OKA has also been
while it does not induce oxidative DNA damage in HepG2 cells and shown to produce condensation of chromatin, reorganization of
showed that OKA induces both cytotoxicity and genotoxicity, cytoskeleton, and DNA fragmentation characteristic of apoptosis
including DNA strand breaks and oxidative DNA damage, in the (Boe et al., 1991; Fernandez-Sanchez et al., 1996). It has been
cells evaluated (Valdiglesias et al., 2011). Protein phosphatase reported to OKA cause neuronal cell death in vitro (Cagnoli et al.,
inhibitor OKA induced protein phosphorylation is an important 1996) and in vivo (He et al., 2001; Yi et al., 2005) by inhibition of
mechanism for the regulation of DNA-PK protein kinase activity the serine/threonine phosphatases 1 and 2A (Cohen, 1990). OKA
and that the protein phosphatase responsible for reactivation in may have rapid metabolic consequences leading to cell death by
vivo is a PP2A-like enzyme (Douglas et al., 2001). However, the altering rates of phosphorylation–dephosphorylation in vivo (Arias
extents of these effects are cell type dependent Reactive nitrogen et al., 1998). Loss of cortical pyramidal neurons, synapse loss,
species (RNS) and reactive oxygen species (ROS) tirggers neuronal (DeKosky and Scheff, 1990) and reduced glutamate concentration,
damage via activation of microglia (Liu et al., 2003). OKA induced together with the formation of neurofibrillary tangles, (Wilcock
cell damage in neuronal cells and evaluated the effects of OKA as et al., 1982) all correlate with the symptoms of dementia. OKA
changes in the quantity of lipid peroxidation products, protein treated rats showed vacuoles, shrinkage, cell blebbing and
carbonyl groups, reduced glutathione content (GSH), glutathione sponginess in cortical and hippocampal areas which indicate
peroxidases (GSH-Px), superoxide dismutase (SDS), catalase, free neurodegeneration (Kamat et al., 2011). A neurite network was
radical generation in rat brain and total lactate dehydrogenase generated in a main channel through the neurite outgrowth from
(LDH) (Tunez et al., 2006). both cell compartments (Kunze et al., 2011). This communication
presents a novel experimental model for Alzheimer studies, where
3.5. Neuroinflammation connected primary neurons were set into subtend, co-pathological
states. Silver staining and immunohistochemistry staining
Neuropathological studies have revealed the presence of a broad revealed that OKA treatment induces NFTs-like conformational
variety of inflammation-related proteins (complement factors, changes in both the cortex and hippocampus (Zhang and Simpkins,
acute-phase proteins and pro-inflammatory cytokines) in AD brains. 2010a,b). Interestingly, mice expressing a dominant negative form
Neuroinflammatory mediators and generation of both reactive of PP2A in neurons display features of Alzheimer’s pathology (Kins
oxygen species (ROS) and reactive nitrogen species (RNS), which et al., 2003). Wang et al. (2004) reported that OKA-induced
affect synapses and induce neuronal damage (Rai et al., 2012). Glial declines in cell viability and mitochondrial metabolic activity
cells activated by amyloid-beta deposits and these cells secrete along with hyperphosphorylation/accumulation of neurofilament-
inflammatory mediators and reactive oxygen species, which can (NF-) H/M subunits. Li et al. (2006) propose that the absence of
aggravate the aggregation of amyloid-beta. Some of the products cleavage and degradation of hyperphosphorylated tau (due to
released by activated glial cells, as well as amyloid-beta itself, can PP2A inhibition) may lead to its accumulation in degenerating
induce or promote neurodegeneration (Hoozemans et al., 2002). neurons. Axonal swellings filled with vesicles, Cell blebbing,
These constituents of innate immunity are involved in several sponginess and changed morphology of cell were observed in OKA
crucial pathogenic events of the underlying pathological cascade in treated rat brain (Kamat et al., 2011) OKA treatment caused axonal
AD, and recent studies have shown that innate immunity is involved swellings filled with vesicles microtubule fragments, and transport
in the etiology of late-onset AD (Eikelenboom et al., 2000). molecules such as kinesin and synapsin and immunocytochemis-
Neuropathological and experimental studies indicate that fibrillar try of the APP C-terminus showed that APP accumulated in the
amyloid-b (Ab) can activate the innate immunity-related CD14 and axonal swellings (Yoon et al., 2006). It has reported that inhibition
Toll-like receptor signaling pathways of glial cells for pro- of protein phosphatases increases tau phosphorylation and
inflammatory cytokine production (Eikelenboom et al., 2010). The initiates neuronal cell death which, include altered Ca2+ homeo-
activation of microglia releases proinflammatory cytokines tumor stasis and glutamate excitotoxicity (Butterfield and Pocernich,
necrosis factor-alpha (TNF-a) and interleukin-beta (IL1-b) (Tyagi 2003). OKA induced cell death via increased reactive oxygen
et al., 2008). Several reports suggested that pro inflammatory species, protein carbonylation, lipid peroxidation, caspase-3
cytokines, oxygen and nitrogen centered free radicals contribute to activity and mitochondrial dysfunction in rat brain (Kamat
the neurodegenerative processes (Tanaka et al., 2006). TNF-a and et al., 2011). It has also reported that OKA offer neuroprotective
IL1-b are suggested as an important mediator in brain pathology of effects against MPP(+)-induced apoptosis by blocking ROS
AD (Tan et al., 2007). OKA administration in rat brain caused elevated stimulation and ROS-mediated signaling pathways in SH-SY5Y
level of cytokine like tumor necrosis factor alpha (TNF-a) and cells indicated that OKA could provide a therapeutic strategy for
interleukin-1 b (IL-1b) and their release caused neuroinflammation the treatment of neurodegenerative diseases (Ahn et al., 2009).
(Kamat et al., 2011). Clinical studies suggest that peripheral Glutamate plays an essential role in learning and memory by
inflammation increases the risk of dementia, especially in patients triggering NMDA receptors to let a controlled amount of calcium
with preexistent cognitive impairment, and accelerates further into a nerve cell. The calcium helps creates the chemical
deterioration in demented patients. OKA injection into the CA1 environment required for information storage. Excess glutamate,
region of the rat hippocampus caused increased in GFAP positive on the other hand, over stimulates NMDA receptors so that they
astrocytes indicates glial activation. allow too much calcium into nerve cells that leads to disruption
and death of cells (Kamat et al., 2010).
3.6. OKA induced neurotoxicity and cell death
4. Concluding remarks
OKA has been shown to be cytotoxic in a variety of cell lines and
Serine/threonine protein phosphatases are important mediators of In conclusion, Protein phosphorylation having wider implica-
general cellular function as well as neurodegenerative processes. tion in learning and memory, Therefore, OKA may be the suitable
OKA increases phosphorylation of microtubule associated protein alternative model to understand the mechanism underlying the
and tau, which are concomitant with early changes in neuronal process of neurodegeneration linked with memory deficit and AD
170 P.K. Kamat et al. / NeuroToxicology 37 (2013) 163–172

pathology. OKA is known to deteriorate memory in elderly person. Bennett PC, Moutsoulas P, Lawen A, Perini E, Ng KT. Novel effects on memory observed
following unilateral intracranial administration of okadaic acid, cyclosporin A,
Therefore from translational point of view, OKA induced memory FK506 and [MeVal4]CyA. Brain Res 2003;988(1–2):56–68.
impairment model may have advantage over the models based on Bialojan C, Takai A. Inhibitory effect of a marine- sponge toxin, okadaic acid, on protein
neurotoxins used in animals or genetic manipulation. There is very phosphatases. Biochem J 1988;256:283–90.
Binder LI, Guillozet-Bongaarts AL, Garcia-Sierra F, Berry RW. Tau, tangles, and Alzhei-
low incidence of genetic based dementia. Such a model may also be mer’s disease. Biochim Biophys Acta 2005;1739(2–3):216–23.
capable to detect the drug acting simultaneously on protein Blass JP, Baker AC, Ko L, Black RS. Induction of Alzheimer antigens by an uncoupler of
phosphorylation and memory process. Such a drug will be of oxidative phosphorylation. Arch Neurol 1990;47(8):864–9.
Bliss TV, Collingridge GL. A synaptic model of memory: long-term potentiation in the
immense important senile dementia of AD type where hyper hippocampus. Nature 1993;361(6407):31–9.
phosphorylation of protein are immediately associated with Boe R, Gjertsen BT, Vintermyr OK, Houge G, Lanotte M, Doskeland SO. The protein
cognitive dysfunction. OKA leads to phosphorylation of tau and phosphatase inhibitor okadaic acid induces morphological changes typical of
apoptosis in mammalian cells. Exp Cell Res 1991;195(1):237–46.
GSK-3b by inhibition of PP-2A activity. OKA also encourage the
Butterfield DA, Pocernich CB. The glutamatergic system and Alzheimer’s disease:
extracellular deposits of b-amyloid in senile plaques, intracellular therapeutic implications. CNS Drugs 2003;17:641–52.
formation of NFT (consisting a phosphorylated form of a Cagnoli CM, Kharlamov E, Atabay C, Uz T, Manev H. Apoptosis induced in neuronal
microtubule associated protein, tau), and the loss of neuronal cultures by either the phosphatase inhibitor okadaic acid or the kinase inhibitor
staurosporine is attenuated by isoquinolinesulfonamides H-7, H-8, and H-9. J Mol
synapses and pyramidal neurons. OKA also persuades oxidative Neurosci 1996;7(1):65–76.
stress, mitochondrial dysfunction, cholinergic dysfunction altered Capetillo-Zarate E, Gracia L, Tampellini D, Gouras GK. Intraneuronal Ab accumulation,
NMDARs function, apoptotic cell death, activate inflammatory amyloid plaques, and synapse pathology in Alzheimer’s disease. Neurodegener Dis
2012;10(1–4):56–9.
cascade and neurotoxicity. All these altered functions are Cardoso SM, Proenca MT, Santos S, Santana I, Oliveira CR. Cytochrome c oxidase is
contributory factors for development of AD like pathology. It is decreased in Alzheimer’s disease platelets. Neurobiol Aging 2004a;25(1):105–10.
evident from the shared information that AD like pathology Cardoso SM, Santana I, Swerdlow RH, Oliveira CR. Mitochondria dysfunction of
Alzheimer’s disease cybrids enhances Abeta toxicity. J Neurochem
induced by OKA builds up a similar condition like clinical 2004b;89(6):1417–26.
neuropathology of AD. Chopra K, Misra S, Kuhad A. Neurobiological aspects of Alzheimer’s disease. Expert
Opin Ther Targets 2011;15(5):535–55.
Clarke NA, Webster M, Francis PT, Procter AW, Hodgkiss AD, Bowen DM. b-Amyloid
5. Future prospectus precursor protein-like immunoreactivity can be altered in humans by drugs
affecting neurotransmitter function. Neurodegeneration 1993;2:243–8.
There are many neurotoxins used as a tool to study AD Cohen P. The structure and regulation of protein phosphatases. Adv Second Messenger
Phosphoprotein Res 1990;24:230–5.
pathology but none of these are able to target memory deficit Collingridge GL, Isaac JT, Wang YT. Receptor trafficking and synaptic plasticity. Nat Rev
through PP2A inhibition (hyperphosphorylation of tau protein). Neurosci 2004;5(12):952–62.
Their targets are likes oxidative stress and glucose metabolism by 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
STZ, MAP protein by Colchicine’s, Ca++ by Kainic acid, and the okadaic acid-induced model of dementia. Behav Brain Res 2012;226(2):420–7.
cholinergic neuron by AF64. In present scenario AChE inhibitors Cui H, Kong Y, Zhang H. Oxidative stress, mitochondrial dysfunction, and aging. J Signal
and NMDA receptor antagonist drugs are the main stay of AD Transduct 2012;646354.
Dalley JW, Theobald DE, Berry D, Milstein JA, Laane K, Everitt BJ, et al. Cognitive
treatment. Since tau hyperphosphorylation is a main pathological
sequelae of intravenous amphetamine self-administration in rats: evidence for
hallmark of AD pathology. None of the drugs are available, which is selective effects on attentional performance. Neuropsychopharmacology
acting through inhibition of tau hyperphosphorylation. So, we 2005;30(3):525–37.
DeKosky ST, Scheff SW. Synapse loss in frontal cortex biopsies in Alzheimer’s disease:
suggest that OKA will be an emerging alternative suitable tool to
correlation with cognitive severity. Ann Neurol 1990;27(5):457–64.
search a therapeutic approach for AD pathology. Del Barrio L, Martin-de-Saavedra MD, Romero A, Parada E, Egea J, Avila J, et al.
Neurotoxicity induced by okadaic acid in the human neuroblastoma SH-SY5Y line
can be differentially prevented by alpha7 and beta2* nicotinic stimulation. Toxicol
Conflict of interest Sci 2011;123(1):193–205.
Dickey CA, Kamal A, Lundgren K, Klosak N, Bailey RM, Dunmore J, et al. The high-
Authors have no conflict of interest. affinity HSP90-CHIP complex recognizes and selectively degrades phosphorylated
tau client proteins. J Clin Invest 2007;117(3):648–58.
Dolan PJ, Johnson GV. The role of tau kinases in Alzheimer’s disease. Curr Opin Drug
Acknowledgement Discov Devel 2010;13(5):595–603.
Douglas P, Moorhead GB, Ye R, Lees-Miller SP. Protein phosphatases regulate DNA-
dependent protein kinase activity. J Biol Chem 2001;276(22):18992–8.
Financial support to Shivika Rai and Pradip Kumar Kamat is Eikelenboom P, Rozemuller AJ, Hoozemans JJ, Veerhuis R, van Gool WA. Neuroin-
gratefully acknowledged to Council of Scientific and Industrial flammation and Alzheimer disease: clinical and therapeutic implications. Alzhei-
Research (CSIR) New Delhi, India. mer Dis Assoc Disord 2000;14(Suppl. 1):S54–61.
Eikelenboom P, van Exel E, Hoozemans JJ, Veerhuis R, Rozemuller AJ, van Gool WA.
Neuroinflammation – an early event in both the history and pathogenesis of
References Alzheimer’s disease. Neurodegener Dis 2010;7(1–3):38–41.
Emmerling MR, Spiegel K, Watson MD. Inhibiting the formation of classical C3-
Abraha A, Ghoshal N, Gamblin TC, Cryns V, Berry RW, Kuret J, et al. C-terminal convertase on the Alzheimer’s beta-amyloid peptide. Immunopharmacology
inhibition of tau assembly in vitro and in Alzheimer’s disease. J Cell Sci 2000;113(Pt 1997;38(1–2):101–9.
21):3737–45. Evans DB, Rank KB, Bhattacharya K, Thomsen DR, Gurney ME, Sharma SK. Tau
Ahn KH, Kim YS, Kim SY, Huh Y, Park C, Jeong JW. Okadaic acid protects human phosphorylation at serine 396 and serine 404 by human recombinant tau protein
neuroblastoma SH-SY5Y cells from 1-methyl-4-phenylpyridinium ion-induced ap- kinase II inhibits tau’s ability to promote microtubule assembly. J Biol Chem
optosis. Neurosci Lett 2009;449(2):93–7. 2000;275(32):24977–83.
Arias C, Sharma N, Davies P, Shafit-Zagardo B. Okadaic acid induces early changes in Fernandez MT, Zitko V, Gascon S, Novelli A. The marine toxin okadaic acid is a potent
microtubule-associated protein 2 and tau phosphorylation prior to neurodegen- neurotoxin for cultured cerebellar neurons. Life Sci 1991;49(19):PL157–62.
eration in cultured cortical neurons. J Neurochem 1993;61(2):673–82. Fernandez JJ, Candenas ML, Souto ML, Trujillo MM, Norte M. Okadaic acid, useful tool
Arias C, Becerra-Garcia F, Arrieta I, Tapia R. The protein phosphatase inhibitor okadaic for studying cellular processes. Curr Med Chem 2002;9(2):229–62.
acid induces heat shock protein expression and neurodegeneration in rat hippocam- Fernandez-Sanchez MT, Garcia-Rodriguez A, Diaz-Trelles R, Novelli A. Inhibition of
pus in vivo. Exp Neurol 1998;153(2):242–54. protein phosphatases induces IGF-1-blocked neurotrophin-insensitive neuronal
Ballatore C, Lee VM, Trojanowski JQ. Tau-mediated neurodegeneration in Alzheimer’s apoptosis. FEBS Lett 1996;398(1):106–12.
disease and related disorders. Nat Rev Neurosci 2007;8(9):663–72. Francis PT. Pyramidal neurone modulation: a therapeutic target for Alzheimer’s
Ballatore C, Brunden KR, Huryn DM, Trojanowski JQ, Lee VM, Smith AB III. Microtubule disease. Neurodegeneration 1996;5(4):461–5.
stabilizing agents as potential treatment for Alzheimer’s disease and related Francis PT, Palmer AM, Snape M, Wilcock GK. The cholinergic hypothesis of Alzheimer’s
neurodegenerative tauopathies. J Med Chem 2012;55(21):8979–96. disease: a review of progress. J Neurol Neurosurg Psychiatry 1999;66(2):137–47.
Bennett PC, Zhao W, Ng KT. Concentration-dependent effects of protein phosphatase Garcia C, Gonzalez V, Cornejo C, Palma-Fleming H, Lagos N. First evidence of dino-
(PP) inhibitors implicate PP1 and PP2A in different stages of memory formation. physistoxin-1 ester and carcinogenic polycyclic aromatic hydrocarbons in smoked
Neurobiol Learn Mem 2001;75(1):91–110. bivalves collected in the Patagonia fjords. Toxicon 2004;43:121–31.
P.K. Kamat et al. / NeuroToxicology 37 (2013) 163–172 171

Gendron TF, Petrucelli L. The role of tau in neurodegeneration. Mol Neurodegener Lu Y, Rosenberg PA. NMDA receptor-mediated extracellular adenosine accumulation is
2009;4:13. blocked by phosphatase 1/2A inhibitors. Brain Res 2007;1155:116–24.
Gong CX, Singh TJ, Grundke-Iqbal I, Iqbal K. Phosphoprotein phosphatase activities in Maidana M, Carlis V, Galhardi FG, Yunes JS, Geracitano LA, Monserrat JM, et al. Effects
Alzheimer disease brain. J Neurochem 1993;61(3):921–7. of microcystins over short- and long-term memory and oxidative stress generation
Gong CX, Lidsky T, Wegiel J, Zuck L, Grundke-Iqbal I, Iqbal K. Phosphorylation of in hippocampus of rats. Chem Biol Interact 2006;159(3):223–34.
microtubule-associated protein tau is regulated by protein phosphatase 2A in Martin L, Latypova X, Terro F. Post-translational modifications of tau protein: implica-
mammalian brain, implications for neurofibrillary degeneration in Alzheimer’s tions for Alzheimer’s disease. Neurochem Int 2011;58(4):458–71.
disease. J Biol Chem 2000;275(8):5535–44. Mather M, Rottenberg H. Aging enhances the activation of the permeability transition
Gong CX, Wang JZ, Iqbal K, Grundke-Iqbal I. Inhibition of protein phosphatase 2A pore in mitochondria. Biochem Biophys Res Commun 2000;273(2):603–8.
induces phosphorylation and accumulation of neurofilaments in metabolically Matias WG, Creppy EE. Evidence for enterohepatic circulation of okadaic acid in mice.
active rat brain slices. Neurosci Lett 2003;340(2):107–10. Toxic Subst Mech 1996b;15:405–14.
Hampel H, Bürger K, Teipel SJ, Bokde AL, Zetterberg H, Blennow K. Core candidate Matsuo ES, Shin RW, Billingsley ML, Van deVoorde A, O’Connor M, Trojanowski JQ, et al.
neurochemical and imaging biomarkers of Alzheimer’s disease. Alzheimers Biopsy-derived adult human brain tau is phosphorylated at many of the same sites
Dement 2008;4(1):38–48. as Alzheimer’s disease paired helical filament tau. Neuron 1994;13(4):989–1002.
He J, Yamada K, Zou LB, Nabeshima T. Spatial memory deficit and neurodegeneration Moreira PI, Zhu X, Wang X, Lee HG, Nunomura A, Petersen RB, et al. Mitochondria: a
induced by the direct injection of okadaic acid into the hippocampus in rats. therapeutic target in neurodegeneration. Biochim Biophys Acta 2010;1802(1):
J Neural Transm 2001;108(12):1435–43. 212–20.
He J, Yang Y, Xu H, Zhang X, Li XM. Olanzapine attenuates the okadaic acid-induced Murakami T, Kobayashi T, Terasawa T, Ohnishi M, Kato S, Sasahara Y, et al. Characteri-
spatial memory impairment and hippocampal cell death in rats. Neuropsycho- zation of multiple molecular forms of Mg(2+)-dependent protein phosphatase
pharmacology 2005;30(8):1511–20. from Saccharomyces cerevisiae. J Biochem 1994;115(4):762–6.
Hirai T, Nohara R, Ogoh S, Chen LG, Kataoka K, Li XH, et al. Serial evaluation of fatty acid Natalia CB, Clara T, Fred VL. Protein tau: prime cause of synaptic and neuronal
metabolism in rats with myocardial infarction by pinhole SPECT. J Nucl Cardiol degeneration in Alzheimer’s disease. Int J Alzheimers Dis 2012. http://
2001;8(4):472–81. dx.doi.org/10.1155/2012/251426.
Hoozemans JJ, Veerhuis R, Rozemuller AJ, Eikelenboom P. The pathological cascade of Nicholls DG, Johnson-Cadwell L, Vesce S, Jekabsons M, Yadava N. Bioenergetics of
Alzheimer’s disease: the role of inflammation and its therapeutic implications. mitochondria in cultured neurons and their role in glutamate excitotoxicity.
Drugs Today (Barc) 2002;38(6):429–43. J Neurosci Res 2007;85(15):3206–12.
Huganir RL, Greengard P. Regulation of neurotransmitter receptor desensitization by Nishimura T, Takeda M, Nakamura Y, Yosbida Y, Arai H, Sasaki H, et al. Basic and clinical
protein phosphorylation. Neuron 1990;5(5):555–67. studies on the measurement of tau protein in cerebrospinal fluid as a biological
Kamat PK, Tota S, Saxena G, Shukla R, Nath C. Okadaic acid (ICV) induced memory marker for Alzheimer’s disease and related disorders: multicenter study in Japan.
impairment in rats: a suitable experimental model to test anti-dementia activity. Methods Find Exp Clin Pharmacol 1998;20(3):227–35.
Brain Res 2010;1309:66–74. Nitsch RM. From acetylcholine to amyloid: neurotransmitters and the pathology of
Kamat PK, Tota S, Shukla R, Ali S, Najmi AK, Nath C. Mitochondrial dysfunction: a crucial Alzheimer’s disease. Neurodegeneration 1996;5(4):477–82.
event in okadaic acid (ICV) induced memory impairment and apoptotic cell death Nunomura A, Tamaoki T, Tanaka K, Motohashi N, Nakamura M, Hayashi T, et al.
in rat brain. Pharmacol Biochem Behav 2011;100(2):311–9. Intraneuronal amyloid beta accumulation and oxidative damage to nucleic acids in
Kamat PK, Tota S, Rai S, Swarnkar S, Shukla R, Nath C. A study on neuroinflammatory Alzheimer disease. Neurobiol Dis 2010;37(3):731–7.
marker in brain areas of okadaic acid (ICV) induced memory impaired rats. Life Sci Palmer AM, Procter AW, Stratmann GC, Bowen DM. Excitatory amino acid-releasing and
2012a;90(19–20):713–20. cholinergic neurones in Alzheimer’s disease. Neurosci Lett 1986;66(2):199–204.
Kamat PK, Tota S, Rai S, Shukla R, Ali S, Najmi AK, et al. Okadaic acid induced Paudel HK. The regulatory Ser262 of microtubule-associated protein tau is phosphor-
neurotoxicity leads to central cholinergic dysfunction in rats. Eur J Pharmacol ylated by phosphorylase kinase. J Biol Chem 1997;272(3):1777–85.
2012b;690(1–3):90–8. Pera M, Alcolea D, Sanchez-Valle R, Guardia-Laguarta C, Colom-Cadena M, Badiola N, et
Kamat PK, Rai S, Swarnkar S, Shukla R, Ali S, Najmi AK, Nath C. Okadaic acid-induced tau al. Distinct patterns of APP processing in the CNS in autosomal-dominant and
phosphorylation in rat brain: role of NMDA receptor. Neuroscience 2013. http:// sporadic Alzheimer disease. Acta Neuropathol 2013;125(2):201–13.
dx.doi.org/10.1016/j.neuroscience.2013.01.075 in press. Perez-Otano I, Ehlers MD. Learning from NMDA receptor trafficking: clues to the
Kar S, Issa AM, Seto D, Auld DS, Collier B, Quirion R. Amyloid beta-peptide inhibits high- development and maturation of glutamatergic synapses. Neurosignals 2004;
affinity choline uptake and acetylcholine release in rat hippocampal slices. 13(4):175–89.
J Neurochem 1998;70(5):2179–87. Poppek D, Keck S, Ermak G, Jung T, Stolzing A, Ullrich O, et al. Phosphorylation inhibits
Kar S, Slowikowski SP, Westaway D, Mount HT. Interactions between beta-amyloid and turnover of the tau protein by the proteasome: influence of RCAN1 and oxidative
central cholinergic neurons: implications for Alzheimer’s disease. J Psychiatry stress. Biochem J 2006;400(3):511–20.
Neurosci 2004;29(6):427–41. Qian W, Shi J, Yin X, Iqbal K, Grundke-Iqbal I, Gong CX, et al. PP2A regulates tau
Kins S, Kurosinski P, Nitsch RM, Gotz J. Activation of the ERK and JNK signaling phosphorylation directly and also indirectly via activating GSK-3beta. J Alzheimers
pathways caused by neuron-specific inhibition of PP2A in transgenic mice. Am J Dis 2010;19(4):1221–9.
Pathol 2003;163(3):833–43. Radebaugh TS, Buckholtz N, Khachaturian Z. Behavioral approaches to the treatment of
Koss DJ, Hindley KP, Riedel G, Platt B. Modulation of hippocampal calcium signalling Alzheimer’s disease: research strategies. Int Psychogeriatr 1996;8(Suppl. 1):7–12.
and plasticity by serine/threonine protein phosphatases. J Neurochem 2007; Rahman A, Akterin S, Flores-Morales A, Crisby M, Kivipelto M, Schultzberg M, et al.
102(4):1009–23. High cholesterol diet induces tau hyperphosphorylation in apolipoprotein E defi-
Kunze A, Meissner R, Brando S, Renaud P. Co-pathological connected primary neurons cient mice. FEBS Lett 2005;579(28):6411–6.
in a microfluidic device for Alzheimer studies. Biotechnol Bioeng 2011;108(9): Rai S, Kamat PK, Nath C, Shukla R. A study on neuroinflammation and NMDA receptor
2241–5. function in STZ (ICV) induced memory impaired rats. J Neuroimmunol 2012 pii:
Li L, Sengupta A, Haque N, Grundke-Iqbal I, Iqbal K. Memantine inhibits and reverses S0165-5728(12)00252-4.
the Alzheimer type abnormal hyperphosphorylation of tau and associated neu- Reynolds CH, Garwood CJ, Wray S, Price C, Kellie S, Perera T, et al. Phosphorylation
rodegeneration. FEBS Lett 2004a;566(1–3):261–9. regulates tau interactions with Src homology 3 domains of phosphatidylinositol 3-
Li X, An WL, Alafuzoff I, Soininen H, Winblad B, Pei JJ. Phosphorylated eukaryotic kinase, phospholipase Cgamma1, Grb2, and Src family kinases. J Biol Chem
translation factor 4E is elevated in Alzheimer brain. Neuroreport 2004b;15(14): 2008;283(26):18177–86.
2237–40. Sadot E, Gurwitz D, Barg J, Behar L, Ginzburg I, Fisher A. Activation of m1 muscarinic
Li X, Lu F, Wang JZ, Gong CX. Concurrent alterations of O-GlcNAcylation and phosphor- acetylcholine receptor regulates tau phosphorylation in transfected PC12 cells.
ylation of tau in mouse brains during fasting. Eur J Neurosci 2006;23(8):2078–86. J Neurochem 1996;66(2):877–80.
Lim YW, Yoon SY, Choi JE, Kim SM, Lee HS, Choe H, et al. Maintained activity of Sas K, Robotka H, Toldi J, Vecsei L. Mitochondria, metabolic disturbances, oxidative
glycogen synthase kinase-3beta despite of its phosphorylation at serine-9 in stress and the kynurenine system, with focus on neurodegenerative disorders.
okadaic acid-induced neurodegenerative model. Biochem Biophys Res Commun J Neurol Sci 2007;257(1–2):221–39.
2010;395(2):207–12. Sergeant N, Bretteville A, Hamdane M, Caillet-Boudin ML, Grognet P, Bombois S, et al.
Lin H, Vicini S, Hsu FC, Doshi S, Takano H, Coulter DA, et al. Axonal alpha7 nicotinic ACh Biochemistry of tau in Alzheimer’s disease and related neurological disorders.
receptors modulate presynaptic NMDA receptor expression and structural plas- Expert Rev Proteomics 2008;5(2):207–24.
ticity of glutamatergic presynaptic boutons. Proc Natl Acad Sci U S A 2012;107(38): Shin DS, Wilkie MP, Pamenter ME, Buck LT. Calcium and protein phosphatase 1/2A
16661–6. attenuate N-methyl-D-aspartate receptor activity in the anoxic turtle cortex. Comp
Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative Biochem Physiol A Mol Integr Physiol 2005;142(September (1)):50–7.
diseases. Nature 2006;443(7113):787–95. Sueoka E, Sueoka N, Okabe S, Kozu T, Ohta T, Suganuma M, et al. Expression of the
Liu SJ, Wang JZ. Alzheimer-like tau phosphorylation induced by wortmannin in vivo tumor necrosis factor alpha gene and early response genes by nodularin, a liver
and its attenuation by melatonin. Acta Pharmacol Sin 2002;23(2):183–7. tumor promoter in primary cultured rat hepatocytes. Cancer Res Clin Oncol
Liu W, Akhand AA, Takeda K, Kawamoto Y, Itoigawa M, Kato M, et al. Protein 1997;123:413–9.
phosphatase 2A-linked and -unlinked caspase-dependent pathways for down- Suuronen T, Kolehmainen P, Salminen A. Protective effect of L-deprenyl against
regulation of Akt kinase triggered by 4-hydroxynonenal. Cell Death Differ apoptosis induced by okadaic acid in cultured neuronal cells. Biochem Pharmacol
2003;10(7):772–81. 2000;59(12):1589–95.
Lovestone S, Reynolds CH, Latimer D, Davis DR, Anderton BH, Gallo JM, et al. Takai A, Bialojan C, Troschka M, Ruegg JC. Smooth muscle phosphatase inhibition and
Alzheimer’s disease-like phosphorylation of the microtubule-associated protein force enhancement by block sponge toxin. FEBS Lett 1987;217:81–4.
tau by glycogen synthase kinase-3 in transfected mammalian cells. Curr Biol Takashima A. GSK-3 is essential in the pathogenesis of Alzheimer’s disease. J Alzhei-
1994;4(12):1077–86. mers Dis 2006;9(3 Suppl.):309–17.
172 P.K. Kamat et al. / NeuroToxicology 37 (2013) 163–172

Tanaka S, Fakher M, Barbour SE, Schenkein HA, Tew JG. Influence of proinflammatory Wilcock GK, Esiri MM, Bowen DM, Smith CC. Alzheimer’s disease, correlation of cortical
cytokines on Actinobacillus actinomycetemcomitans specific IgG responses. choline acetyltransferase activity with the severity of dementia and histological
J Periodontal Res 2006;41(1):1–9. abnormalities. J Neurol Sci 1982;57(2–3):407–17.
Tessier S, Godreau D, Vranckx R, Lang-Lazdunski L, Mercadier JJ, Hatem SN. Cumulative Wu S, Sasaki A, Yoshimoto R, Kawahara Y, Manabe T, Kataoka K, et al. Neural stem cells
inactivation of the outward potassium current: a likely mechanism underlying improve learning and memory in rats with Alzheimer’s disease. Pathobiology
electrical memory in human atrial myocytes. J Mol Cell Cardiol 2001;33(4): 2008;75(3):186–94.
755–67. Yamaguchi R, Perkins G. Dynamics of mitochondrial structure during apoptosis and the
Thiels E, Kanterewicz BI, Knapp LT, Barrionuevo G, Klann E. Protein phosphatase- enigma of Opa1. Biochim Biophys Acta 2009;1787(8):963–72.
mediated regulation of protein kinase C during long-term depression in the adult Yi KD, Simpkins JW. Protein phosphatase 1, protein phosphatase 2A, and calcineurin play a
hippocampus in vivo. J Neurosci 2000;20(19):7199–207. role in estrogen-mediated neuroprotection. Endocrinology 2008;149(10):5235–43.
Tian Q, Lin ZQ, Wang XC, Chen J, Wang Q, Gong CX, et al. Injection of okadaic acid into Yi KD, Chung J, Pang P, Simpkins JW. Role of protein phosphatases in estrogen-
the meynert nucleus basalis of rat brain induces decreased acetylcholine level and mediated neuroprotection. J Neurosci 2005;25(31):7191–8.
spatial memory deficit. Neuroscience 2004;126(2):277–84. Yi KD, Covey DF, Simpkins JW. Mechanism of okadaic acid-induced neuronal death and
Torrent L, Ferrer I. PP2A and Alzheimer disease. Curr Alzheimer Res 2012;9(2):248–56. the effect of estrogens. J Neurochem 2009;108(3):732–40.
Túnez I, Collado JA, Medina FJ, Muñoz MC, Gordillo R, Sampedro C, Moyano MJ, Feijóo Yin YY, Liu H, Cong XB, Liu Z, Wang Q, Wang JZ, et al. Acetyl-L-carnitine attenuates
M, Muntané J, Montilla P. Protective effect of carvedilol on oxidative stress induced okadaic acid induced tau hyperphosphorylation and spatial memory impairment
by okadaic acid in N1E-115 cells. Pharmacol Res 2006;54(3):241–6. in rats. J Alzheimers Dis 2010;19(2):735–46.
Tyagi E, Agrawal R, Nath C, Shukla R. Influence of LPS-induced neuroinflammation Yoon SY, Choi JE, Yoon JH, Huh JW, Kim DH. BACE inhibitor reduces APP-beta-C-
on acetylcholinesterase activity in rat brain. J Neuroimmunol 2008;205(1–2): terminal fragment accumulation in axonal swellings of okadaic acid-induced
51–6. neurodegeneration. Neurobiol Dis 2006;22(2):435–44.
Valdiglesias V, Laffon B, Pasaro E, Cemeli E, Anderson D, Mendez J. Induction of Zhang Y, Herman B. Apoptosis and successful aging. Mech Ageing Dev 2002;123(6):
oxidative DNA damage by the marine toxin okadaic acid depends on human cell 563–5.
type. Toxicon 2011;57(6):882–8. Zhang Z, Simpkins JW. Okadaic acid induces tau phosphorylation in SH-SY5Y cells in an
van de Nes JA, Konermann S, Nafe R, Swaab DF. Beta-protein/A4 deposits are not estrogen-preventable manner. Brain Res 2010a;1345:176–81.
associated with hyperphosphorylated tau in somatostatin neurons in the hy- Zhang Z, Simpkins JW. An okadaic acid-induced model of tauopathy and cognitive
pothalamus of Alzheimer’s disease patients. Acta Neuropathol 2006;111(2): deficiency. Brain Res 2010b;1359:233–46.
126–38. Zhang J, Cheng Y, Zhang JT. Protective effect of ( ) clausenamide against neurotoxicity
Wagner U, Utton M, Gallo JM, Miller CC. Cellular phosphorylation of tau by GSK-3 beta induced by okadaic acid and beta-amyloid peptide25-3. Yao Xue Xue Bao
influences tau binding to microtubules and microtubule organisation. J Cell Sci 2007;42(9):935–42.
1996;109(Pt 6):1537–43. Zhao WQ, Sedman GL, Gibbs ME, Ng KT. Effect of PKC inhibitors and activators on
Wang J, Tung YC, Wang Y, Li XT, Iqbal K, Grundke-Iqbal I. Hyperphosphorylation and memory. Behav Brain Res 1994;60(2):151–60.
accumulation of neurofilament proteins in Alzheimer disease brain and in okadaic Zheng L, Kågedal K, Dehvari N, Benedikz E, Cowburn R, Marcusson J, et al. Oxidative
acid-treated SY5Y cells. FEBS Lett 2001;507(1):81–7. stress induces macroautophagy of amyloid beta-protein and ensuing apoptosis.
Wang DL, Ling ZQ, Cao FY, Zhu LQ, Wang JZ. Melatonin attenuates isoproterenol- Free Radicl Biol Med 2009;46:422–9.
induced protein kinase A overactivation and tau hyperphosphorylation in rat brain. Zhou XW, Gustafsson JA, Tanila H, Bjorkdahl C, Liu R, Winblad B, et al. Tau hyperpho-
J Pineal Res 2004;37(1):11–6. sphorylation correlates with reduced methylation of protein phosphatase 2A.
Wang Z, Yang L, Zheng H. Role of APP and Abeta in synaptic physiology. Curr Alzheimer Neurobiol Dis 2008;31(3):386–94.
Res 2012;9(2):217–26. Zimmer ER, Kalinine E, Haas CB, Torrez VR, Souza DO, Muller AP, et al. Pretreatment with
Whitehouse PJ, Kellar KJ. Nicotinic and muscarinic cholinergic receptors in Alzheimer’s memantine prevents Alzheimer-like alterations induced by intrahippocampal oka-
disease and related disorders. J Neural Transm Suppl 1987;24:175–82. daic acid administration in rats. Curr Alzheimer Res 2012 Dec;9(10):1182–90.

You might also like