Abstract
Current psychiatric diagnostic schema segregate symptom clusters into discrete entities, however, large proportions of patients suffer from comorbid conditions that fit neither diagnostic nor therapeutic schema. Similarly, psychotropic treatments ranging from lithium and antipsychotics to serotonin reuptake inhibitors (SSRIs) and acetylcholinesterase inhibitors have been shown to be efficacious in a wide spectrum of psychiatric disorders ranging from autism, schizophrenia (SZ), depression, and bipolar disorder (BD) to Alzheimer’s disease (AD). This apparent lack of specificity suggests that psychiatric symptoms as well as treatments may share aspects of pathophysiology and mechanisms of action that defy current symptom-based diagnostic and neuron-based therapeutic schema.
A myelin-centered model of human brain function can help integrate these incongruities and provide novel insights into disease etiologies and treatment mechanisms. Available data are integrated herein to suggest that widely used psychotropic treatments ranging from antipsychotics and antidepressants to lithium and electroconvulsive therapy share complex signaling pathways such as Akt and glycogen synthase kinase-3 (GSK3) that affect myelination, its plasticity, and repair. These signaling pathways respond to neurotransmitters, neurotrophins, hormones, and nutrition, underlie intricate neuroglial communications, and may substantially contribute to the mechanisms of action and wide spectra of efficacy of current therapeutics by promoting myelination. Imaging and genetic technologies make it possible to safely and noninvasively test these hypotheses directly in humans and can help guide clinical trial efforts designed to correct myelination abnormalities. Such efforts may provide insights into novel avenues for treatment and prevention of some of the most prevalent and devastating human diseases.
Pervasive brain myelination underlies neural network synchrony and our distinctiveness as a species. Psychiatric diagnoses may share deficits in myelin development, plasticity, or repair. Treatments act on neuroglial signaling pathways such as Akt and GSK3 that improve myelination. Treatment efficacy may derive from myelination-driven improved neural network synchronization. “Neuroglialpharmacology” encapsulates a paradigm shift in medication development strategy.
Keywords: White matter, oligodendrocyte, intracortical myelin, medication, MRI, NG2 cells, neuregulin, ErbB, DISC1, IGF1, Reelin, Cdk, MAPK, mTOR, Leptin
1. Introduction
The diagnostic schema embodied in the Diagnostic and Statistical Manual of Mental Disorders (DSM) is symptom-based and largely disconnected from disease etiologies or a biologic model of healthy human brain function. Useful models of brain function, as well as the dysfunctions that manifest in psychopathology, must by necessity incorporate testable hypotheses that help explain both the mode of action of psychotropic medications as well as their wide spectrum of efficacy (e.g., ability to treat multiple diagnoses). With notable exceptions (e.g., lithium, electroconvulsive treatments, and nutritional interventions), much of current clinical pharmacology is believed to act primarily at neuronal synapses that are largely confined to gray matter. This has focused clinical and research attention on neurons and synapses residing therein. This narrow focus has contributed to the under-appreciation of glia and especially oligodendrocytes and the myelin sheaths they produce in optimizing the timing and synchrony of action potentials on which optimal function of neuronal networks depends. Timing is a key metric to all cortical operations (Klausberger and Somogyi, 2008) and it is primarily dependent on the production, maintenance, and repair of myelin (reviewed in Bartzokis et al., 2011d).
The human brain is exceptionally myelinated compared to other species (Smaers et al., 2011) (reviewed in Bartzokis et al., 2011d). This extensive myelination has imposed exceptionally high metabolic demands and is associated with vulnerabilities that make the human species highly susceptible to distinctive and highly prevalent brain disorders throughout its lifespan (reviewed in Bartzokis, 2002, 2004, 2011a; Bartzokis et al., 2011d). While most think of myelin as a component of white matter, in humans, gray matter is also extensively myelinated (Figure 1), and the key role of this intracortical myelin (ICM) component in optimizing brain function have generally been overlooked.
Figure 1. Quadratic (inverted “U”) Myelination Trajectories of Human Brain Over the Lifespan.
Human brain myelination (Y axis) across the age span (X axis). Frontal and temporal lobes (depicted on left and right figures respectively). Top figures are in vivo MRI data (from Bartzokis et al., 2001) showing significant quadratic relationships (p<.001) of myelinated white matter volumes (a measure that includes highly myelinated lower cortical layers). Intracortical myelin stain data are depicted in the lower figures (from Kaes, 1907) (adapted and reproduced in Kemper, 1994). Used with permission. The in vivo (top panels) and post-mortem (lower panels) samples of normal individuals show remarkably similar myelination trajectories demonstrating a dynamic myelin “plasticity” that does not peak until middle age. Note: even though the regions are similar, as is the case with these two late-myelinating association regions, myelination trajectories differ and peak at significantly different ages (p<.01) (Bartzokis et al., 2001).
This report will focus on two important aspects of brain function and dysfunction from the perspective of myelin. First, the importance of intracortical myelin to the plasticity required to continually optimize the timing of action potentials and network oscillations on which learning, cognitive performance, and behavior depend (sections 2–4). Second, the shared mechanisms of action of psychotropic treatments on brain myelin and its continual optimization and repair (sections 5 and 6). These underappreciated aspects of myelination may help explain the efficacy that different classes of pharmaceuticals have on the same sets of symptoms as well as the wide spectrum of efficacy of certain classes of medications on multiple diagnoses/symptom clusters. The emphasis herein on a myelin perspective is not meant to deny the important role of synaptic activity in psychiatric disease and its treatment, rather, the aim is to balance and integrate that neuronal perspective with the key role of glia and myelin in particular in normal and abnormal brain function.
2. Myelin and the Human Brain
The brain is classically divided into gray matter (defined as the regions containing neuronal cell bodies and almost all synaptic connections) and white matter (composed primarily of the very long neuronal appendage (axon) that acts as a “wire” connecting widely dispersed neurons, plus the oligodendrocytes that produce the axon’s “insulating” myelin sheaths). The roughly 100 billion neurons in the human brain are actually a small minority of brain cells (10%) while glia, which are present in both gray and white matter, account for the rest: astrocytes (45%), oligodendrocytes (35%), microglia (5%), and progenitor (NG2) cells (5%, the vast majority of which differentiate into oligodendrocytes). The human brain consumes 20% of our total energy expenditure compared to 13% in monkeys and 2-8% in other vertebrates. This striking shift in resource use was made possible by important evolutionary adaptations in lipid and energy metabolism. Compared to other species, these adaptations made it possible to devote a greater proportion (approximately 25%) of our brain’s mass to myelin and thus achieve the information processing capacity that defines the human species (reviewed in Bartzokis, 2011b).
Human brain myelination has a quadratic-like (inverted “U”) trajectory across the lifespan with increasing myelin content that peaks in middle–age (Figure 1). The “connectivity” provided by myelination consists of increased action potential transmission speed (over 100-fold) and decreased refractory time (34-fold) which increases the number of action potentials that can be transmitted per unit time (in Internet terminology this would represent expanded “bandwidth”). Myelination thus potentially increases the information processing capacity of our brain’s “Internet” by over 3,000 fold, making human myelination indispensable for developing our species’ elaborate cognitive functions (reviewed in Bartzokis et al., 2011d). Human cognitive functions are also highly dependent on later-myelinating oligodendrocytes. These cells myelinate the circuitry of our neural networks all the way to the neuron bodies located in gray matter structures such as the cortex (Figure 1). The extensive intracortical myelination process occurs later (after childhood) and basically “upgrades” neural networks with immediate response capacity such that they are essentially “on line” and process information much more quickly and precisely (reviewed in Bartzokis, 2011b) (see section 3.1).
Although the brain is routinely conceptualized as a singular entity, it is composed of a myriad of interacting neural networks that have highly “plastic” and dynamic developmental and degenerative trajectories. As Figure 1 demonstrates, even at the gross lobar level, the different trajectories reach peak myelination at different ages. These different trajectories are supported by oligodendrocytes that become increasingly more complex the later in life they differentiate. They range from robust oligodendrocytes that myelinate a single axon segment with over 100 wraps of myelin membrane in the early-myelinating motor and sensory regions/networks to more vulnerable oligodendrocytes that myelinate as many as 50 axon segments with less than 10 wraps in late-myelinating intracortical regions (Figure 1) (reviewed in Butt and Berry, 2000). The structurally more complex and metabolically overextended later-myelinating oligodendrocytes and their myelin are especially vulnerable during both developmental (Stark et al., 2004; Uranova et al., 2004; Vostrikov and Uranova, 2011; Vostrikov et al., 2007) as well as degenerative phases (Bartzokis et al., 2004; Marner et al., 2003) of the myelination trajectories over the lifespan (reviewed in Bartzokis, 2011b).
From the perspective of the exceptionally myelinated human species, the development and maintenance/repair of myelin’s functional integrity may be the single most important and vulnerable element for acquiring and maintaining optimal cognitive and behavioral function. In short, myelin may arguably represent the “weakest link” of both brain development as well as age-related degeneration and thus contribute to many of the normal as well disease-causing changes in brain function over the entire human lifespan. Including glia and myelin in a “model” of the human brain can help explain normal brain function, clinical and pathologic phenomenology of multiple diseases, as well as their shared responsiveness to pharmaceutical and other (e.g., nutritional) interventions (section 5 and 6).
3. Function of the Brain’s Biologic “Internet” is Directly Dependent on Myelin
3.1 Normal/Optimal Brain Function Depends on Synchrony of Oscillations
Brain regions communicate through synchronized firing of populations of neurons in networks whose activity is reflected in the extracellular field potential as brain oscillations. Oscillations can be measured through techniques such as electroencephalography (Williams and Boksa, 2010). Oscillation-based synchrony is the most energy-efficient physical mechanism for temporal coordination. Mammalian cortical neurons form behavior-dependent oscillating networks that span a very wide frequency spectrum (from 0.05 to 500 Hz) creating tremendous information processing potential. These networks vary in size, are phylogenetically conserved, and oscillation-based functions they support can be highly heritable (Hong et al., 2008; Linkenkaer-Hansen et al., 2007) (reviewed in Buzsaki and Draguhn, 2004). Synchronized oscillations may also establish the precise action potential timing needed for use-dependent synaptic plasticity (e.g., long term potentiation) to occur (Rutishauser et al., 2010) (reviewed in Uhlhaas and Singer, 2010). Conversely, asynchronous arrival of action potentials has been shown to contribute to synaptic loss/pruning (Purves and Lichtman, 1980).
The first step towards network synchronization is achieved in childhood by myelinating the subcortical white matter portion of axons connecting widely distributed brain regions into functional networks (Figure 2). This initial subcortical myelination can be initiated/directed by neuronal signals themselves (Fields and Burnstock, 2006; Ziskin et al., 2007) (reviewed in Butt, 2011) and results in the remarkably faster conduction (>100 times faster than unmyelinated axon) between widely separated gray matter regions such as thalamus and the many cortical regions with which it interacts. Once subcortical myelination is achieved, the total conduction time between these highly dispersed regions becomes primarily dependent on the much longer time (roughly 10 times) action potentials spend traversing the short but unmyelinated portion of axons within cortex. This intracortical distance to a specific neuronal layer is roughly constant. The constant intracortical distance to layer III (which receives most of the cortico-cortical input), together with the slow intracortical signal propagation, establishes the initial roughly synchronous arrival of action potentials to all cortical regions that are at different distances (Kimura and Itami, 2009; Salami et al., 2003). The rough network synchrony achieved by this process underlines the vast repertoire of cognitive and behavioral abilities that can be achieved in childhood albeit few of these functions or their integration are “perfected”/optimized at that early stage of life (Bartzokis, 2011b).
Figure 2. The Human Brain “Internet” Establishes Synchronous Activity in Childhood Through Subcortical Myelination and Perfects it in Adulthood Through Intracortical Myelin (ICM).
Red arrows stand in for myelinated axons with fast transmission (more than 100 fold faster than unmyelinated axons). Insert depicts cortex of child (on left, unmyelinated) and adult (on right, myelinated). Subcortical myelination (accomplished in childhood) makes action potential transmission in circuits of markedly different lengths such as thalamo-frontal (long horizontal red arrow) and thalamoinsular (short down-pointing red arrow) very fast. Once the fast subcortical portion of a circuit is myelinated, the intracortical transmission of the action potential through unmyelinated axons (insert: blue arrow on left panel) takes much longer (10 fold longer than subcortical transmission). Thus, once subcortical myelination is established in childhood, the constant intracortical distance to layer III establishes a roughly synchronous arrival of action potentials to all layer III pyramidal neurons. Later in development and throughout adulthood, appropriate and region-specific myelination of the cortical portion of the axons (insert: red arrow on right panel) continually optimizes the synchrony of action potential arrival and thus optimizes cognitive and behavioral functions (see text for further details).
3.2 Intracortical Myelin (ICM) Optimizes Network Oscillations and Brain Function
As described above, the short intracortical portion of axonal propagation (that is largely unmyelinated in childhood) exerts a markedly disproportionate influence on synchronicity of action potential arrival across functional networks and their vast numbers of neurons and synapses. Beyond childhood, even faster transmission as well as exquisitely more precisely synchronized timing can be achieved by adding the appropriate amounts of myelin to the intracortical portion of fibers. As Figure 1 suggests, cortical oligodendrogenesis occurs primarily in adulthood (O'Kusky and Colonnier, 1982) and underlies the acceleration and “fine grained” synchronization of cognitive and behavioral networks that continue to be refined over the entire first six decades of life. This later-differentiating intracortical subgroup of oligodendrocytes seems to differ in subtle ways from their subcortical counterparts (Kessaris et al., 2006; Noble et al., 2003; Power et al., 2002), as may the composition of the myelin they produce (Hartman et al., 1982) (reviewed in Butt and Berry, 2000). Cortical myelination underlies a key mechanism of brain plasticity and its disturbance could have important consequences for disease pathophysiology as well as efficacy of psychotropic treatments (Bartzokis, 2011b; Bartzokis et al., 2009) (see section 4, and 5).
Myelin-based network plasticity is dependent on continued oligogenesis (Figure 1). Life-long oligogenesis is a distinctive oligodendrocyte feature that is central to brain development and plasticity throughout life. Unlike neurons, whose numbers are essentially established at birth, in healthy primates, vast numbers of progenitor (NG2) cells are produced to support the decades-long processes of postnatal myelination and repair/remyelination (Levine et al., 2001). The NG2 cells comprise approximately 5% of total adult brain cells and continue to divide, increasing the number of differentiated oligodendrocytes by as much as 50% during adulthood (O'Kusky and Colonnier, 1982; Peters and Sethares, 2004; Peters et al., 2008; Vostrikov and Uranova, 2011; Vostrikov et al., 2007). By dividing and differentiating into oligodendrocytes, NG2 cells can support both continued myelination of additional axons or portions thereof (e.g., intracortical) as well as remyelinate damaged or lost myelin sheaths (Peters and Sethares, 2003; Peters et al., 2008). The plasticity of intracortical myelin (ICM) could also compensate for network synchrony disruptions brought by changes in transmission speeds anywhere in the circuitry, including those resulting from subcortical myelin repair processes that can alter transmission speed by decreasing myelin thickness (Smith and Koles, 1970) (reviewed in Bartzokis, 2011b) (Figure 2).
Although there are multiple possible causes for pathologic changes in circuit oscillations, the importance of ICM in compensating for subcortical transmission delays and optimizing brain function is supported by observations from multiple sclerosis (MS), a canonical myelin disease, and Alzheimer’s disease (AD), usually considered a canonical “cortical” disease. Until recently myelin-destroying intracortical MS lesions, which postmortem data show represent as much as 60% of MS lesions, were under-appreciated due in part to difficulty in detecting them on MRI (reviewed in Simon et al., 2010). Prospective studies show that absence of such cortical lesions is associated with a favorable clinical and cognitive outcome independent of deep white matter lesion accumulation (Calabrese et al., 2009). Conversely, the presence and progression of intracortical lesions in MS are most clearly associated with cognitive decline (including processing speed and memory) (Roosendaal et al., 2009). These phenomena can be parsimoniously explained by the plasticity of ICM and its ability to compensate for subcortical delays in transmission and re-establishing network synchrony. Thus, only when the optimizing role of ICM is lost to intracortical demyelination would subcortical delays fully manifest as degraded network synchrony and function and thus become observable as clinical symptoms. Similar focal losses of intracortical myelin associated with amyloid beta (Aβ) plaques were recently documented in AD (Mitew et al., 2010) and may similarly contribute to declines in cognitive and behavioral functions observed in that disease, although this possibility has only recently begun to be directly investigated in vivo (Bartzokis et al, unpublished data).
4. Dysregulated Myelination in Schizophrenia (SZ) and Bipolar Disorder (BD)
Over the last decade the importance of myelin pathology in SZ and BD has become widely recognized (reviewed in Andreasen et al., 2011; Bartzokis, 2002, 2011b; Brambilla et al., 2009; Corfas et al., 2004; Davis et al., 2003; Dwork et al., 2007; McIntosh et al., 2009). Although white matter abnormalities are present in both diseases, the patterns of abnormalities are not identical (Bartzokis, 2011b).
In chronic SZ, post-mortem gene expression, cytology, and myelin stain studies provide converging evidence to support the view of a deficient trajectory of frontal lobe ICM. Imaging studies that assessed white matter volume provided converging evidence of a deficient myelination trajectory that, unlike in healthy individuals (Figure 1), ceases its development during early adulthood. Similar oligodendrocyte reductions and myelin gene expression deficits are also observed in chronic BD and may even occur in chronic severe unipolar depression (reviewed in Bartzokis, 2011b).
The data on disease-related changes in earlier-myelinating subcortical white matter is more complex and may differ in SZ and BD. In SZ, the bulk of post mortem studies suggest that subcortical myelin deficits are absent or not as prominent as cortical myelin/oligodendrocyte defects and imaging studies examining subcortical white matter of younger groups of SZ subjects (mean age 26 years or younger) using DTI also suggest that abnormalities are not present at disease onset but rather develop as the disease progresses (Friedman et al., 2008; White et al., 2011) (reviewed in Bartzokis, 2011b). A recent post mortem study supports this apparent progression of subcortical white matter involvement with disease durations. It showed that subcortical myelin defects are observed almost exclusively in brains of older SZ subjects, are associated with longer durations of illness, and are limited to earlier-myelinating large and medium size fibers (Uranova et al., 2011). A trajectory of progressive subcortical myelin/white matter disruption may also be reflected in DTI data from studies that assessed older-onset first-episode SZ subjects (mean onset age >26 years), which generally reported significant deficits in white matter integrity (reviewed in Bartzokis, 2011b). These differences may be influenced by a greater repair potential of subcortical white matter (Power et al., 2002) and by age-related reductions in myelin repair potential (Shen et al., 2008).
The thinner myelin produced by remyelination (Peters, 2009) slows conduction (Smith and Koles, 1970) and may thus contribute to degradation of network synchrony. The intracortical myelination processes observed in healthy controls (Figure 1) seems to be deficient in chronic SZ as well as BD (Bartzokis et al., 2011b; Vostrikov and Uranova, 2011) and therefore, compensating for subcortical changes in conduction velocity may be inadequate or fail altogether (Bartzokis, 2011b). Inadequate control of intracortical myelination could eventually degrade the synchrony of neural network oscillations (see section 3.2) and result in cognitive and behavioral inefficiencies and disorganization that are part of the clinical manifestations of several psychiatric disorders (Bartzokis, 2011b).
Compared to SZ, in BD subcortical myelin deficits can be more prominent (Regenold et al., 2007) and on MRI, focal regions of subcortical myelin damage (manifest as increased signal intensity in white matter) is consistently reported in BD (Altshuler et al., 1995) (reviewed in Brambilla et al., 2009; Kempton et al., 2008). Thus, in contrast to SZ where initially ICM deficits may be most prominent, in BP disorder increased vulnerability of earlier-myelinating subcortical fibers may be more pronounced at disease onset. More efficient repair mechanisms of subcortical myelin (Power et al., 2002) would permit the reestablishment of network synchrony and recovery of function, and may be aided by treatments such as lithium (Brambilla et al., 2009; van der Schot et al., 2009). This suggests that in BD adequate ICM plasticity may initially be able to compensate for subcortical transmission delays in BD to a greater extent than in SZ. Nevertheless, post-mortem data suggest that as BD progresses into its chronic phases, significant intracortical oligodendrocyte deficits develop in BD as they do in SZ (Rajkowska et al., 2001; Uranova et al., 2004; Vostrikov and Uranova, 2011). These ICM deficits may help account for the eventual appearance of cognitive deficits and functional decline in chronic BD (Burdick et al., 2011; Sole et al., 2011) despite cognitive abilities in youth that may be above average, in contrast to SZ where cognitive deficits are present at onset (Koenen et al., 2009; Zanelli et al., 2010).
5. Psychotropic Treatments Influence Glia and Myelination
Activity-dependent neuroglial communication can be supported through neuronal ATP release as well as its metabolite adenosine. ATP activates purinergic receptors that modulate intracellular calcium and cyclic AMP and have multiple effects on glia, oligodendrocytes, and myelination (Butt, 2011; Fields and Burnstock, 2006). In addition, all the major neurotransmitter systems on which the bulk of currently available psychotropic medications act (dopamine, serotonin, norepinephrine, acetylcholine, GABA, and glutamate) may have important roles in myelination. Neurotransmitter-based neuroglial communication can influence/direct myelination and is supported by at least three mechanisms that will be reviewed next: synaptic, extra-synaptic, and non-synaptic. Their influence on myelination/repair processes may be especially important in synapse-rich cortical and other gray matter regions where all these neurotransmitter-based mechanisms can operate jointly (Bartzokis, 2007; Belachew et al., 1999; Butt, 2006; Karadottir and Attwell, 2007).
5.1 Synaptic, Extra-Synaptic, and Non-Synaptic Neurotransmitter Effects on Glia
Neuronal glutaminergic and GABAergic synapses onto oligodendrocyte progenitors (NG2 cells) have been demonstrated in both the developing brain and in white matter undergoing remyelination following experimental myelin damage (Etxeberria et al., 2010; Velez-Fort et al., 2010) (reviewed in Kolodziejczyk et al., 2010; Mangin and Gallo, 2011). Such direct neurotransmitter-based neuroglial communication mechanisms may have functional importance in oligodendrocyte differentiation and myelin repair as indicated by in vitro work showing an influence of both AMPA type glutamate receptors and GABA-A receptors on migration and differentiation (Gudz et al., 2006; Tong et al., 2009; Yuan et al., 1998).
In addition to direct synapses, neuroglial signaling may also occur through extra-synaptic transmission (also called volume transmission) due to “spillover” of neurotransmitters from synapses or nodes of Ranvier. This neuroglial signaling mechanism may be especially significant during high-frequency discharges and oscillations that release larger volumes of neurotransmitters. The direct synapses that GABA interneurons form onto NG2 cells in development seem to be converted into this kind of extra-synaptic GABA-oligodendrocyte transmission during later maturation (Velez-Fort et al., 2010). Thus, extra-synaptic neuroglial communication mechanisms may be especially important for the plasticity needed to optimize the oscillation synchrony and timing of highfrequency networks that are best supported by myelinated axons (Bartzokis et al., 2010) (see section 3).
Multiple classes of existing psychotropic treatments (e.g., antipsychotics, antidepressants) target neurotransmission and have substantial yet underappreciated neuroglial signaling roles. A very large proportion of cholinergic transmission (up to 90%) both in the developing and adult brain is non-synaptic (e.g., absence of post-synaptic neuronal button), with acetylcholine being released from cholinergic varicosities directly into the extracellular space (reviewed in Bartzokis, 2007). In addition to acetylcholine, catecholamines (primarily dopamine, serotonin, and norepinephrine) are also largely (>50–80%) non-synaptically released (Descarries et al., 1996; Parent et al., 2010; Smiley et al., 1994; Umbriaco et al., 1995) (reviewed in Fuxe and Agnati, 1991). These non-synaptic and extra-synaptic neuroglial communications can impact oligodendrocyte differentiation and myelination (reviewed in Bartzokis, 2007, 2011b).
It is of interest to note that glia may also influence neurotransmitter-based extra- and non-synaptic signaling through secretion of most of the extracellular matrix components such as reelin (see section 5.2.1) and chondroitin sulfate proteoglycans. This extracellular matrix differs from healthy controls in SZ but not BD and could contribute to some of the differences in clinical manifestations (reviewed in Berretta, 2011) despite shared myelination deficits between diseases (see section 4). Such glial-dependent influences would add another level of control as well as complexity to neuroglial communication through diffusible signaling molecules such as neurotransmitters. The following sections will integrate therapeutic as well as countertherapeutic (from drugs of abuse) influences on myelin plasticity from dopaminergic, serotinergic, GABAergic, glutaminergic, and cholinergic signaling (section 5.2.2 and 5.2.3), as well as growth factor, neurotrophic, hormonal (section 6.2), and nutritional factors (section 6.3).
5.2 Multiple Psychotropic Medications Inhibit Glycogen Synthase Kinase 3 (GSK3) and May Promote Myelination
The human species’ exceptional myelination is supported by very recent evolutionary changes involving apolipoprotein E, lactate dehydrogenase, and peroxisome organelle function. These adaptations may have evolved in part to support the extremely metabolically “expensive” processes of creating and maintaining a highly myelinated CNS (Rinholm et al., 2011). Thus, metabolic derangements that would have relatively subtle sequelae peripherally, may produce significant dysfunction in brain. It is thus not surprising that metabolic abnormalities such as insulin resistance and brain lipidation seem to increase AD risk, predate the onset of psychiatric disease such as schizophrenia and bipolar disorder, and are associated with worse outcomes (reviewed in Bartzokis, 2011b). Given the very recent evolution of myelinating oligodendrocytes (in vertebrates), myelination’s exceptional metabolic requirements had to be integrated with the many metabolic and developmental processes that predated its evolution.
Glycogen synthetase kinase 3 (GSK3) (and other kinases that have similar/overlapping functions (see section 6.1)) is highly conserved from sponges, through insects and vertebrates (Adamska et al., 2011). By the time myelin evolved, many processes were already modulated by GSK3 through its >40 substrates that include metabolic and signaling proteins, structural proteins, and transcription factors in different cellular compartments such as within cytoplasm, and also in nucleus and mitochondria where GSK3 is highly active. The integration of these other functions with GSK3 effects on myelination (see next section) may have further increased the complexity of GSK3 actions and contributed to the plethora of pharmacologic and non-pharmacologic interventions that may impact the myelination process (Figure 3) (Jope and Johnson, 2004; Sutherland, 2011). As will be reviewed below, neurotransmitter-based as well as non-neurotransmitter-based effects on myelination include many different classes of psychotropic treatments ranging from lithium and other mood stabilizers, to antipsychotics, acetylcholinesterase inhibitors, serotonin reuptake inhibitors (SSRIs), and electroconvulsive treatments (ECT). It is therefore proposed that the efficacy of much of the current clinical pharmacology and therapeutics may be due, at least in part, to treatment-induced changes in glia and in particular oligodendrocytes and their myelin (section 2 and 3). This concept gave rise to and is embodied in the term neuroglial pharmacology (reviewed in Bartzokis, 2011b). The remaining subsections (5.2.1 – 5.2.3) and section 7 will review some of the signaling mechanisms influencing myelination.
Figure 3. Neuroglialpharmacology of Psychiatric Treatments, Hormones, and Neurotrophins.
Major classes of currently available psychotropic medications, other pharmacologic agents, and ECT (underlined) seem to share direct or indirect inhibition of GSK3 as a common mechanism of action. Many but not all GSK3 effects are shared by its α and β isoforms, however in many instances, specific isoform effects remain to be clarified. Indirect (through Akt activation) and direct GSK3β inhibition promotes myelination by releasing the negative control GSK3β has on myelination. Some neurotrophins (NRG & BDNF), hormones (IGF1, T3, TRH), and cytokines (leptin) also seem to inhibit GSK3. Conversely, some drugs of abuse with known cognitive and behavioral toxicities (cocaine, methamphetamine, PCP) seem to have the opposite (GSK3 activation) effect and would be expected to share inhibition of myelination as a potential detrimental effect on brain function. Some GSK3-dependent myelin interactions of the dopaminergic, serotinergic, glutaminergic, and cholinergic system are depicted in this figure, however the GABAergic ones are not (see text for details). An asterisk denotes genes known to be associated with increased risk for schizophrenia (SZ) and/or bipolar disorder (BD).
Note: as a schematic focused on the key role of GSK3 on myelination, this figure does not depict many additional relationships such as the ones between GSK3 and other kinases (mTOR, MAPK, Cdk), genes, nutrition, metabolism, environment, and epigenetic changes and the interdependence of all CNS cell types and their specialized structures such as synapses. For further details on these topics please refer to the text and reference list.
5.2.1 The Akt/GSK3 Myelination Signaling Pathway
Many important classes of psychotropic medications seem to share a mechanism involving Akt (also known as protein kinase B) and GSK3 that are at the core of a signaling cascade with multiple inputs as well as downstream effects (Figure 3). Akt is a serine/threonine kinase regulated through phosphatidylinositol 3- kinase (PI3K)-mediated signaling that is conserved in vertebrates (Souza et al., 2011). Akt exists in three isoforms (Akt1 – ubiquitously expressed, Akt2 – predominantly expressed in insulin target tissues, and Akt3 – predominant isoform in brain) that show strong homology but are coded by different genes. Akt can phosphorylate GSK3β at the serine-9 position and GSK3α at the serine-21 position and thus inhibit their activity (reviewed in Sutherland, 2011). Historically, GSK3 was associated with glycogen synthesis in response to insulin. It also exists in two closely related isoforms (α and β) coded on different genes (Figure 3).
GSK3 is an unusual serine/threonine kinase that is constitutively active and is primarily controlled by inhibition. Furthermore, GSK3 preferentially (by 100–1000 fold) phosphorylates pre-primed (pre-phosphorylated) substrates (Thomas et al., 1999) and has over 40 substrates ranging from metabolic and signaling proteins to structural proteins and transcription factors. Other kinases can thus influence GSK3 signaling directly of indirectly, by pre-phosphorylating its substrates (see section 6.1). GSK3 is thus a point of convergence and acceleration (for pre-phosphorylated substrates) for multiple signaling pathways (reviewed in Beaulieu and Gainetdinov, 2011; Jope and Johnson, 2004; Sutherland, 2011; Wang et al., 2011b). The GSK3 isoforms (α and β) have overlapping but not identical substrates as illustrated by the apparent specificity of GSK3a activation in promoting amyloid beta (Aβ) protein production while GSK3β activation promotes tau protein phosphorylation (reviewed in Zhao and Townsend, 2009). For many substrates however, the amount of overlap in activity between GSK3α and β isoforms has not been fully elucidated (Polter et al., 2010).
In addition to its other functions in energy production, inflammation, and apoptosis (Sutherland, 2011), GSK3β has been shown to be a powerful negative regulator of oligodendrocyte differentiation and myelination (Figure 3) that can override the effects of other pathways such as Wnt signaling by controlling multiple regulators (Azim and Butt, 2011). Active GSK3β retards the repopulation of demyelinated axons while its inhibition promotes myelination. At doses achieved in vivo, lithium as well as several other endogenous and exogenous compounds inhibits GSK3β and enhances oligodendrocyte differentiation and myelination without apparent impact on neurons, axons, or astrocytes (Azim and Butt, 2011). Since Akt activation inhibits GSK3 (Figure 3), activators of Akt also have promyelinating effects (Flores et al., 2008; Narayanan et al., 2009) while Akt deficiency can impair prefrontal cortex function and expression of myelin genes (Lai et al., 2006).
The promyelinating effects of the Akt/GSK3 signaling pathway on brain can be substantial. When Akt is driven to be constitutively active, hypermyelination without increasing oligodendrocyte numbers is specifically observed in CNS but not in PNS (Flores et al., 2008; Yu et al., 2011). Conversely, over-expression of GSK3β reduces myelination (Azim and Butt, 2011), brain size, and cortical thickness without a decline in neuron number and thus results in increased neuronal density (Spittaels et al., 2002). This neuronal density increase is similar to increases observed in SZ (Selemon et al., 1995, 1998) that have been suggested to be due, at least in part, to deficient intracortical myelination (Bartzokis, 2002; Bartzokis and Altshuler, 2005). Additional supporting evidence for the role of GSK3 in myelination comes from up-regulating insulin growth factor-1 (IGF1), which also ultimately inhibits GSK3 (Frederick et al., 2007; Ye et al., 2010) (Figure 3) and promotes myelination (Carson et al., 1993; D'Ercole and Ye, 2008; Freude et al., 2008) (see sections 6.1 and 6.2). Conversely, IGF1 deficiency impedes myelination and produces a pattern similar to the ones seen in GSK3 over-expression and SZ consisting of brain atrophy, reduced myelination and cortical thickness, and increased neuronal density without a change in neuronal number (reviewed in Spittaels et al., 2002).
Reelin is another key signaling glycoprotein that is secreted into extracellular matrix, interacts with some of the same receptors as apolipoprotein E, and helps coordinate embryonic and adult brain development and repair (Barr et al., 2007). Reelin interacts with the same signaling pathways as dopamine-2 receptors (DR2 - see next section 5.2.2) and can indirectly inhibit GSK3 (Fatemi et al., 2009) and could thus promote myelination (Figure 3). Conversely, inhibition of reelin should reduce myelination and has been shown to impair cognitive functions (Beffert et al., 2005; Brosda et al., 2011; Stranahan et al., 2011; Wedenoja et al., 2010). Reelin deficits are consistently observed in developmental disorders such as SZ, BD, major depression, and autism (Fatemi et al., 2005a; Fatemi et al., 2005b; Impagnatiello et al., 1998; Torrey et al., 2005) and such deficits could contribute to the myelination deficits observed in these disorders (section 4). Conversely, reelin overexpression seems to prevent behavioral phenotypes related to SZ and BD in animal models (Teixeira et al., 2011). Reelin is secreted by oligodendrocytes and their precursors (Siebert and Osterhout, 2011) and after childhood, it is also secreted by GABAergic interneurons throughout cortical layers II-VI and hippocampus, and may help account for the co-occurrence of reelin and GABA deficits (as well as myelin – see section 4) in psychiatric diseases (Fatemi et al., 2005b; Guidotti et al., 2011; Shen et al., 2008; Torrey et al., 2005).
In striking contrast to developmental disorders associated with reelin deficits, increased reelin is observed in trisomy 21 (Downs) subjects (who all eventually develop degenerative AD pathology) as well as in cognitively normal individuals that nevertheless had AD pathology at post-mortem (Botella-Lopez et al., 2010; Kramer et al., 2011). Conversely, in transgenic mouse models of AD, reduced reelin levels result in accelerated AD pathology (Kocherhans et al., 2010). These observations suggest that in individuals without developmental psychiatric disorders such as SZ and BD, as myelin repair needs increase due to age-related and/or genetic degenerative processes, homeostatic up-regulation of reelin occurs that may inhibit GSK3 and thus promote compensatory remyelination/repair (Bartzokis, 2011a; Kramer et al., 2011). This compensatory up-regulation of reelin seems to be deficient/absent in developmental psychiatric disorders (Fatemi et al., 2005a; Fatemi et al., 2005b; Impagnatiello et al., 1998; Torrey et al., 2005) possibly through epigenetic mechanisms (Guidotti et al., 2011) (see section 6.1) and may help explain the need for exogenous GSK3 inhibition that seems to be provided by so many current therapeutic interventions (Figure 3) (see next section, 5.2.2).
5.2.2 Promyelinating Potential of Major Classes of Psychotropic Medications
Lithium, an inorganic element administered as a salt for the treatment of BD, is a powerful inhibitor of GSK3β. Lithium can inhibit GSK3β directly via competition with magnesium and indirectly by increasing inhibitory serine-phosphorylation of GSK3 through Akt (Li et al., 2010b) (reviewed in Beaulieu and Gainetdinov, 2011; Polter et al., 2010) (Figure 3). Together, these GSK3 inhibitory mechanisms likely mediate the behavioral effects of lithium (Beaulieu et al., 2004) and it is thus possible that myelination is involved in its mechanism of action (Azim and Butt, 2011) (reviewed in Bartzokis, 2011b) (Figure 3). This proposition is indirectly supported by reports that that bipolar susceptibility genes are associated with white matter volume deficits that may be mitigated by treatment with lithium (van der Schot et al., 2009) as well as decreased Akt activity and increased GSK3β activity in the brain of depressed subjects at post-mortem (Karege et al., 2011; Karege et al., 2007) (reviewed in Pan et al., 2011; Polter et al., 2010). Furthermore, lithium treatment seems to up-regulate several myelin proteins including the long isoform of myelin basic protein (McQuillin et al., 2007), and lithium was useful in the treatment, prevention, and reduced recurrence of myelin damage in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS) (De Sarno et al., 2008). Notably however, even though continuous lithium treatment provided long-lasting (90 day) protection from EAE symptoms, withdrawal of lithium resulted in a rapid recurrence of symptoms (De Sarno et al., 2008). This is consistent with the suggestion that continuous inhibition of the constitutively active GSK3β is important for optimal therapeutic effects. In addition, valproic acid, a medication developed for treating seizures that has proven effective in treating BD, also directly inhibits GSB3β (Kim et al., 2005; Miller et al., 2009) (Figure 3) and has promyelinating effects (Fei et al., 2011). The shared GSK3 inhibition of lithium and valproic acid may help explain their shared efficacy in treating BD despite strikingly different molecular structures. The efficacy of typical and atypical antipsychotics in the treatment of BD (reflected in recent FDA approvals of several antipsychotics for BD treatment) may also act through GSK3 inhibition (Figure 3).
As mentioned previously, GSK3β can be inactivated by phosphorylation of a single serine-9 residue by Akt (as well as other kinases – see section 6.1) or indirectly through many activators of Akt (Figure 3). Dopamine 2 receptor (D2R) signaling, is indirectly mediated through a β-arresting 2 (βArr2)/protein phosphatase 2A (PP2A) signaling complex resulting in inactivation of Akt and subsequent activation of GSK3 (Beaulieu et al., 2005). Dopaminergic transmission could thus ultimately inhibit myelination (Figure 3). The long-standing hypothesis that SZ is associated with a hyper dopaminergic state predating the onset of psychosis (Howes et al., 2011; Seeman, 2010) is thus consistent with a dopamine-driven GSK3 activation resulting in the myelination deficits observed in SZ (see section 4). Supporting this possibility are observations that several polymorphisms of enzymes involved in dopaminergic transmission including dopamine metabolism through catechol-Omethyltransferase (COMT), D2R, and Akt are associated with increased risk for psychiatric diseases and/or BD (Blasi et al., 2011) (reviewed in Beaulieu and Gainetdinov, 2011) (Figure 3). Dopamine-induced GSK activation can be overcome by D2R blockade, a property shared by all antipsychotics (Seeman, 2010). Early in treatment, antipsychotics have been shown to promote oligodendrocyte differentiation and myelin repair in rodent models (Wang et al., 2010; Xu et al., 2010), increase cortical glial numbers in primates (Selemon et al., 1999), and increase intracortical myelin in SZ (Bartzokis et al., 2009). These initial effects may contribute to the high levels of symptom remission that are especially striking within the first year of SZ treatment (ranging from 70 to 87%) (Emsley et al., 2007; Lieberman et al., 1993; Nuechterlein et al., 2006; Robinson et al., 2004; Saravanan et al., 2010).
Antipsychotic-induced GSK3 inhibition is short lived however (Li et al., 2007) and medication non-adherence is a well-known problem in psychiatric populations (Tiihonen et al., 2011). Long-acting intramuscular injection (depo) formulations for antipsychotics mitigate adherence problems (Keith, 2009) and have been associated with improved clinical outcomes (Subotnik et al., 2011; Tiihonen et al., 2011) (reviewed in Keith, 2009) possibly by providing continuous inhibition of the constitutively active GSK3. Furthermore, some animal studies suggest that lower doses of antipsychotics may inhibit GSK3 best (Li et al., 2007) and therefore, the troughs and peaks in antipsychotic blood levels associated with the kinetics of oral administration may not be optimal for achieving continuous GSK3 inhibition, as well as possibly increasing risks of untoward side effects. Long-term (> 1 year) treatment with oral antipsychotics has been shown to reduce cortical glial numbers in monkeys (Konopaske et al., 2008). In humans, loss of intracortical oligodendrocytes and myelin is clearly observed at post mortem in SZ subjects after many years of treatment with oral antipsychotics (see section 4) and imaging studies of SZ subjects confirm intracortical myelin deficits in patients chronically treated with oral antipsychotics (Bartzokis et al., 2011b). Whether the decline in intracortical myelin is due to poor adherence, pharmacokinetic factors, the disease process itself, or a combination of these factors remains unclear. Nevertheless, a recent randomized study suggests that, early in the disease course, the trajectory of decline in ICM may be modifiable by continuous treatment with injectable long-acting antipsychotics (Bartzokis et al., 2011a) (Bartzokis et al, unpublished data). The above reports are thus consistent with a recent large study of first-break SZ subjects reporting gray as well as white matter volume losses that were attributed to chronic treatment with oral antipsychotics (Ho et al., 2011) and that white matter (but not gray matter) volume losses were associated with cognitive deterioration (Andreasen et al., 2011), one of the best correlates of clinical outcomes. Thus the poor adherence that often follows remission from the initial SZ episode, could result in dysinhibition of GSK3 and may help explain the decreased myelination and lower white matter volumes as well as the associated cognitive and clinical deterioration that occurs after the first year of treatment (Bartzokis et al., 2011a; Bartzokis et al., 2011b) (reviewed in Bartzokis, 2011b).
As is the case with D2R, activation of D1R and D3R seem to also activate GSK3 (Lebel et al., 2009; Miller et al., 2010; Niu et al., 2010) (reviewed in Beaulieu and Gainetdinov, 2011) and as such, could contribute to myelination deficits observed in SZ and BD (see section 4). This would suggest that blockade of multiple subtypes of dopamine receptors may have promyelinating effects (Figure 3). All antipsychotic medications (typical and atypical) share dopamine receptor blockade however, atypical antipsychotics can also inhibit GSK3 independently of Akt. Atypical antipsychotics differ from typical ones in part by their strong antagonism of serotonin (5HT2A) receptor (5HT2AR). Since 5HT2AR activates GSK3, blocking 5HT2AR would inhibit GSK3 and potentiate the promyelinating effect of D2R blockade (Xu et al., 2010) (reviewed in Li and Jope, 2010) (Figure 3). This additional potential promyelinating effect present only in atypical antipsychotics could help explain a recent observation on antipsychotic-related ICM increases in early stages of treatment. Although both typical and atypical antipsychotics seemed to increase ICM in SZ patients, the atypical one did so to a significantly greater extent (Bartzokis et al., 2009).
Unlike the apparent similar GSK3-activating effects of dopamine acting through several of its receptors, serotonin 5HT2AR and 5HT1AR have opposite effects on GSK3 activity. As reviewed in the prior paragraph, antagonism of 5HT2AR inhibits GSK3 (and thus would increase myelination) while 5HT1AR agonism does the same (Figure 3). Some atypical antipsychotics synergistically combine both these effects (not depicted in Figure 3) and have been reported to especially benefit cognitive function (Meltzer and Massey, 2011). Changes in overall level of brain serotonin can also have significant effects on GSK3 activity. Increased serotonin availability with selective serotonin uptake inhibitors (SSRIs), monoamine oxidase inhibitors, and tricyclic antidepressants have been shown to increase GSK3β inhibition in frontal cortex, hippocampus, and striatum of normal mice and may therefore have promyelinating effects in these brain regions. Conversely, reduced serotonin (in mutants that lack the ability to synthesize it) results in a two-fold increase in GSK3 activity (reviewed in Beaulieu and Gainetdinov, 2011) and would be expected to impair myelination (Azim and Butt, 2011). Interestingly, animal models have shown that additive effects on GSK3 inhibition can be achieved by combining D2R and 5HT2AR blockade (using the atypical antipsychotic risperidone) with monoamine reuptake inhibition (using the SSRI fluoxetine) (Li et al., 2007). This would inhibit GSK3 through D2R plus 5HT2A blockade by risperidone and combine it with additional GSK3 inhibition due to fluoxetine-induced 5HT2 increases that would provide 5HT21AR agonist activity (Figure 3).
The possibility that reduced intracortical myelin in SZ (see section 4) is due to impaired Akt/GSK3 signaling pathway is supported by post-mortem data on SZ frontal cortex showing reduced levels of Akt protein, Akt mRNA, and phosphorylated (inactive) GSK3β (Balu et al., 2010; Emamian et al., 2004; Thiselton et al., 2008; Zhao et al., 2006). Similar findings are reported for mood disorders (Karege et al., 2011; Karege et al., 2007) (reviewed in Polter et al., 2010) and genetic associations between Akt/GSK3 signaling pathway have been reported for both SZ and BD (Karege et al., 2010). Furthermore, cell models and brain structural network function assessed with brain imaging in SZ as well as healthy control subjects demonstrated gene-gene interactions (epistasis) between Akt, PI3K, D2R, and COMT polymorphisms (Blasi et al., 2011; Jagannathan et al., 2010; Nicodemus et al., 2011; Sei et al., 2010) that would be expected from the mechanisms depicted in Figure 3.
In addition to the serotinergic and dopaminergic neurotransmitter effects summarized above, cholinergic stimulation could also influence myelination (Bartzokis, 2007). The mechanism may involve nicotinic α7 receptors that have been shown to inhibit GSK3 (Bitner et al., 2010; Rehani et al., 2008) (Figure 3) and/or muscarinic receptors that indirectly inhibit GSK3 by activating Pi3K/Akt and increase oligodendrocyte precursor survival (Cui et al., 2006) (not depicted in Figure 3). Acetylcholinesterase inhibitors, the current mainstay of AD treatment, reduce acetylcholine breakdown. The resulting increase in acetylcholine levels can stimulate both nicotinic and muscarinic receptors resulting in GSK3 inhibition (De Sarno et al., 2006). These treatments have also been shown to increase IGF-1 levels (Gomez, 2008; Narimatsu et al., 2009; Obermayr et al., 2005) that could indirectly inhibit GSK3 acting through Akt (Figure 3), and may increase white matter volume (Venneri and Lane, 2009). Nicotine and its metabolite cotinine can also stimulate nicotinic α7 receptors and, in addition to possible promyelinating effects, may have anti-inflammatory effects (reviewed in Wang et al., 2011b).
5.2.3 ECT and Adjunctive Treatments May Also Inhibit GSK3 and Promote Myelination
Thyrotropin-releasing hormone (TRH) is a small neuropeptide involved in the hypothalamic-pituitary control of thyroid and other hormones (Wallis, 2010). In addition to canonical effects on gene expression TRH can have more direct and immediate non-genomic effects (Pekary et al., 2006). TRH is widely distributed throughout the brain and has been shown to inhibit GSK3β gene expression (Luo and Stopa, 2004), while GSK3β inhibitors in turn can modulate TRH and TRH-like peptide release (Pekary et al., 2010). Although TRH levels decrease in the hypothalamus in aging rats (Pekary et al., 1984), the levels seem to be preserved in healthy aging humans (Mazzoccoli et al., 2010) however, reduced levels are reported in AD (Luo et al., 2002). TRH can alter cognitive and emotional function (Bennett et al., 1997) and is prominently increased after electroconvulsive treatment (ECT) a widely used clinical intervention that is especially efficacious for severe melancholic and/or psychotic depression (Pekary et al., 1999) (reviewed in Sattin, 1999). ECT may also acutely inhibit GSK3 through the canonical mechanism of Akt activation (Kang et al., 2004; Roh et al., 2003) (Figure 3). ECT has been reported to increase oligogenesis (Wennstrom et al., 2003; Wennstrom et al., 2004), an effect that has also been recently reported with antipsychotics (Niu et al., 2010; Xu et al., 2010) (see section 5.2.2).
Triiodothyronine (T3), the biologically active form of thyroid hormone commonly used as an adjunct in the treatment of depression (Bauer et al., 2005) (reviewed in Bauer et al., 2008), may also inactivate GSK3β by activating the PI3K/Akt cascade (Cao et al., 2009) (Figure 3) and has been shown to regulate oligodendrocyte accumulation in rat white matter tracks (Schoonover et al., 2004). Further support for the promyelinating effects of thyroid hormones comes from the prominent myelination deficits that occur when thyroid deficiency is experienced in development (resulting in cretinism) (Barradas et al., 2001; Pinazo-Duran et al., 2011) as well as deficits in myelin repair efficiency in adulthood (Harsan et al., 2008).
In light of the proposed role for myelin in the pathophysiology of multiple psychiatric disorders and common comorbid manifestations of these disorders (reviewed in Altamura et al., 2011; Bartzokis, 2004, 2005), it should not be surprising that treatment with T3, its pro-hormone T4, or TRH itself have been reported to have antidepressant properties (Bauer et al., 2008; Szuba et al., 2005). Furthermore, several reports suggest that heavily myelinated subcortical fibers are most clearly susceptible to thyroid deficiencies (Barradas et al., 2001; Harsan et al., 2008; Pinazo-Duran et al., 2011). This distribution may help explain the relative specificity of these interventions to mood disorders since subcortical white matter abnormalities seem to be most clearly associated with mood disorders (see section 4) (reviewed in Bartzokis, 2011b).
5.2.4 Drugs of Abuse May Dysregulate Myelination and Result in Psychiatric Symptoms
The prior sections suggests that a major mechanism of action for multiple classes of psychiatric treatments may involve, at least in part, the release of oligodendrocytes and myelination from the negative control of GSK3 (Azim and Butt, 2011). Conversely, increased extracellular dopamine, whether produced by genetic variants that increase risk of psychiatric disease or drugs of abuse such as amphetamine and cocaine, results in GSK3 activation (reviewed in Beaulieu and Gainetdinov, 2011). Elevated extracellular dopamine (in dopamine transporter knock out animal models or administration of amphetamine, methamphetamine or the dopamine agonist apomorphine) has been reported to inhibit Akt and thus activate GSK3 (Bourque et al., 2011) (reviewed in Beaulieu and Gainetdinov, 2011). As expected by the signaling pathways depicted in Figure 3, psychostimulant use has been shown to reduce oligodendrocytes and myelination in susceptible late-myelinating regions such as frontal cortex (Bartzokis et al., 2002; Yang et al., 2011) (reviewed in Tobias et al., 2010). Thus indiscriminate increase of neurotransmitters (e.g., not directly associated with physiologic neurotransmission) caused by drugs of abuse could degrade homeostatic physiologic mechanisms through which neural networks adjust ICM and reestablish network synchrony (see section 5.1). This could undermine the compensatory ICM changes that restore precise timing of action potentials on which optimal function depends. The resulting degradation in network function could secondarily contribute to the cognitive deficits and thought and mood disturbance-inducing effects associated with these drugs of abuse (reviewed in Bartzokis, 2005, 2011b).
Another class of drugs of abuse, N-methyl-D-aspartic acid (NMDA) receptor antagonists such as phencyclidine (PCP) and dizocilpine (MK-801), are also well-known psychosis–inducing (psychotogenic) compounds. They also activate GSK3β by decreasing the phosphorylation/inhibition of Akt (Luo et al., 2003; Xi et al., 2011) (Figure 3). Anticholinergic drugs could have similar deleterious clinical effects (reviewed in Bartzokis, 2011b) by reducing cholinergic inhibition of GSK3 (Figure 3). Thus, different classes of drugs of abuse, acting through different mechanisms (Solowij et al., 2011) yet sharing deleterious effects on cognition and thought and mood control, may share indiscriminate activation of GSK3 as a possible mechanism of action. Conversely, medications that inhibit GSK3, such as D2R and 5HT2A receptor blockers, seem to have therapeutic effects in psychotic disorders whether secondary to drugs of abuse or due to psychiatric disorders.
6.0 Non-Akt/GSK3 Mechanisms Involved in Myelination
Given the complexity, metabolic cost, and functional importance of myelination (Sections 2 and 3), the existence of parallel/redundant mechanisms to control myelination should not be unexpected. Such redundant signaling pathways considerably increase the complexity of phenotypes, however, they also make it possible to integrate/coordinate myelination with the metabolic and hormonal environments as well as neuronal function. Thus, although focused on oligodendrocytes, this article is not meant to suggest that oligodendrocytes are the only target of successful treatments. It does however propose that the production and maintenance of myelin may be the “weakest link” of the human CNS and may represent a common pathophysiology shared amongst multiple neuropsychiatric disorders. The differential involvement of myelin subtypes with different vulnerabilities (e.g. subcortical versus intracortical – see Section 3) may result in different phenotypes despite sharing a common myelin substrate (see Section 4) (Bartzokis, 2011b). This possibility is indirectly supported by the observation that many of the current treatment interventions (e.g., antipsychotics, SSRIs, etc.) have a wide spectrum of efficacy and encompass many disease categories as currently defined in the DSM. This wide spectrum of efficacy suggests that multiple pharmacologic as well as non-pharmacologic interventions may act on a shared myelin vulnerability that, given the exceptionally extensive myelination of the human brain, manifests most distinctly in our species (Bartzokis, 2011b). The existence of a common biological substrate could also explain the complexity of phenotypes and frequent co-existence (comorbidity) of more than one disorder within the same individual (Altamura et al., 2011; Bartzokis, 2004, 2005). The next section will briefly review three additional kinase-based signaling pathways that impact the myelination process and act through overlapping but not identical mechanisms.
6.1 Parallel and/or Redundant Signaling Pathways that Modulate Myelination
Another evolutionarily conserved serine/threonine protein kinase initially identified as a target of the immunosuppressant rapamycin thus named mammalian target of rapamycin (mTOR) can also inhibit GSK3. Mammals have two mTOR complexes, one sensing energy/nutrient status and cellular stress while the other senses primarily growth factors, hormones and cytokines (reviewed in Zoncu et al., 2011). This enzyme may have thus further helped integrate the considerable energy and nutritional needs of oligodendrocytes (see section 2) with the complex signaling (including growth factors) that controls the multiple myelination steps (not all interactions shown in Figure 3). Major roles of mTOR have been established for aging, autophagy (including debris clearance necessary for remyelination), degenerative brain diseases (Garelick and Kennedy, 2011; Spilman et al., 2010) (reviewed in Zoncu et al., 2011), inflammation (Wang et al., 2011a), and myelination (Goebbels et al., 2010; Tyler et al., 2009). It also has complex interactions (crosstalk) with Akt/GSK3 (Garelick and Kennedy, 2011; Wang et al., 2011a; Zoncu et al., 2011) and other signaling pathways (see below). Inhibiting mTOR has been shown to extend lifespan in middle age as well as old rodents (Harrison et al., 2009) and, in transgenic models of AD (a strikingly age-related disease), it seems to decrease cognitive deficits as well as its Aβ and tau pathology (Caccamo et al., 2010). Given some of the multiple interactions between signaling pathways (Figure 3) specific effects are difficult to disentangle (Ma et al., 2010; Spilman et al., 2010) however, increased oligodendrocyte differentiation has been reported with mTOR inhibition (Tyler et al., 2009). In addition to integrating myelination with nutrient and energy status described above, some neurotransmitter signaling mechanisms with antidepressant effects may act through mTOR-dependent mechanisms to integrate myelination with synaptogenesis (Duman et al., 2012; Li et al., 2010a).
Inhibition of GSK3β can also be achieved through two mitogen-activated protein kinase (MAPK) signaling pathways: p38 MAPK and the extracellular signal-regulated kinases 1 and 2 (ERK1/2) (Figure 3). P38 MAPK is activated primarily through cellular stress and cytokines and, unlike Akt, inactivates GSK3β by phosphorylating its C terminus. This parallel pathway is relatively specific to brain, may be specific for activating a cell survival pathway, which is not targeted by the Akt/GSK3 pathway (Fragoso et al., 2007; Thornton et al., 2008), and may be involved in epigenetic modifications of DNA (Day and Sweatt, 2011). The ERK1/2 and p38 pathways have been implicated in peripheral myelination (Fei et al., 2011; Newbern et al., 2011) and CNS oligodendrocyte survival (Althaus and Kloppner, 2006; Du et al., 2006; Kumar et al., 1998), myelination (Haines et al., 2010), and timing of myelination especially in late-myelinating regions (Fyffe-Maricich et al., 2011; Haines et al., 2010; Raff et al., 1988). The MAPK pathways can be triggered by multiple growth factors such as platelet–derived growth factor (PDGF), fibroblast growth factor-2 (FGF-2), nerve growth factor (NGF) (Du et al., 2006), and IGF1 (Bibollet-Bahena and Almazan, 2009) as well as neurotrophins such as brain derived neurotrophic factor (BDNF) and neurotropin-3 (Du et al., 2006; Van't Veer et al., 2009) (reviewed in Althaus et al., 2008; Fyffe-Maricich et al., 2011). These same triggers can also activate the PI3K/Akt pathway (Bibollet-Bahena and Almazan, 2009; Coelho et al., 2009) (Figure 3) and some triggers, such as IGF1, may impact multiple control points in oligodendrocyte survival, proliferation, and differentiation (Bibollet-Bahena and Almazan, 2009) and is thus depicted in Figure 3 by itself as well as subsumed under “growth factors”.
In addition to GSK3, MAPK, and mTOR, a fourth family of protein kinases, cyclin-dependant kinase (Cdk), can impact myelination. Endogenous CNS-specific modifiers of Cdk5 function are altered in SZ brain (Engmann et al., 2011) and can influence myelination (Miyamoto et al., 2007; Tamura et al., 2005). Cdk5 can have dynamic (age-dependent) crosstalk with kinases such as GSK3 (Engmann and Giese, 2009) mediated in part by neuregulin (Wen et al., 2008) and could thus contribute to the age-related decline in myelin repair/remyelination efficiency (Shen et al., 2008) (reviewed in Bartzokis, 2011a). Due to the complexity of Cdk influences (see below) only this aging-dependent influence depicted in Figure 3. The Cdk family is evolutionarily conserved (Hsu et al., 2011) and with the exception of Cdk5 (that in addition to other functions primes GSK3 substrates (Medina and Wandosell, 2011) – see section 5.2.1), several members such as Cdk1, Cdk2, and Cdk4 are involved cell cycle progression. Given that NG2 cells differentiate into oligodendrocytes throughout the lifespan (section 2), it is not surprising that the Cdk family is also directly involved in regulating several aspects of myelination with each member being influenced by different sets of endogenous modifiers (Atanasoski et al., 2008; Caillava et al., 2011; Hsu et al., 2011; Miyamoto et al., 2007). Cdk2 in particular has 45% homology with GSK3 (Lesuisse et al., 2010) and, as is the case with GSK3, inhibition of Cdk2 has recently been shown to accelerate oligodendrocyte precursor differentiation and remyelination in the adult CNS (Caillava et al., 2011). Furthermore, up-regulation of an endogenous Cdk2 inhibitor promotes oligodendrocyte differentiation (Larocque et al., 2005), a process that can be promoted by antidepressants through activation of glucocorticoid receptors (Anacker et al., 2011). Psychotropic medications may thus impact myelination through multiple parallel mechanisms as well as crosstalk between the multiple protein kinases involved in metabolic pathways (Figure 3) that underlie cell cycle progression and differentiation (Frederick and Wood, 2004; Kerns et al., 2010).
6.2 Hormones and Neurotrophins
Akt can inhibit both GSK3α and β in response to multiple hormones and growth factors including BDNF, leptin, IGF1, and insulin itself (Greco et al., 2009, Marwarha et al., 2010) (reviewed in Beaulieu and Gainetdinov, 2011; Polter et al., 2010). The same growth factors can act through parallel pathways involving MAPK and mTOR (see section 6.1) (Figure 3). Thus, at least part of the mechanism of action of these hormones on myelin could be based on reducing the activity of GSK3.
Interactions between the pharmacologic mechanisms reviewed above (section 5.2.2) and the individual’s hormonal state (section 6.1) are also important to consider. Such interactions are suggested by reports that response to acetylcholinesterase inhibitors used in the treatment of AD may be more robust in individuals with higher peripheral levels of IGF1 (Tei et al., 2008), which is normally taken up by the brain from the periphery at rates that surpass those of insulin (Bondy and Cheng, 2004). In addition, treatment interventions themselves may act in part through peripheral mechanisms. For example, antipsychotics have been shown to increase peripheral IGF1 when given to drug-naïve SZ subjects (Venkatasubramanian et al., 2010). Similarly, by increasing peripheral IGF-1 that is taken up by the brain, physical exercise may help improve cognition and mood (Llorens-Martin et al., 2010; Simon et al., 2011; Trivedi et al., 2011) (reviewed in Bartzokis, 2011a). Some oral GSK3 inhibitors have been shown to increase IGF1 transport into brain by interacting with megalin, a major multicargo transport protein that ferries proteins across the blood brain barrier and choroid plexus (Bolos et al., 2011) (Figure 3). The above reports are consistent with studies showing that peripherally infused IGF-1 (and insulin) enter the brain (Aberg et al., 2007; Anderson et al., 2002) through active transport (Yu et al., 2006) and increase cortical oligodendrocytes (Aberg et al., 2007).
6.3 Nutritional Modifiers
Certain nutrients, such as vitamins B12 and folate, seem to have GSK3 inhibitory effects. Conversely, vitamin B12 deficits or resulting hyperhomocysteinemia have also been shown to decrease GSK3 phosphorylation/inhibition (Nicolia et al., 2011; Zhuo et al., 2011) (Figure 3) and could thus impair myelination (Azim and Butt, 2011). This may help explain the epidemiologic (Clarke et al., 1998; Kim et al., 2008) and animal model (Chan and Shea, 2007; Zhuo et al., 2011) studies that report associations between these nutritional deficiencies and increased AD risk as well as white matter deficits (de Lau et al., 2009; Graber et al., 2011) (reviewed in Bartzokis, 2011a).
Omega-3 fatty acids (especially DHA) and iron are also essential nutrients for myelination. Nutritional interventions with these myelin building blocks on oligodendrocytes were reviewed in a prior publication (Bartzokis, 2011b). A subsequent report suggests that a genetic defect in the peroxisome-dependent enzyme that catalyzes the last step of DHA synthesis may put affected individuals at risk for DHA deficiency (Astarita et al., 2010). Individuals with such mutations should be especially helped by DHA supplementation to prevent and possibly treat diseases such as schizophrenia, MDD, autism, and AD where DHA deficits have been reported (reviewed in Bartzokis, 2011b). The contribution of such genetic variants of metabolism to the pathophysiology of these disorders remains to be fully elucidated however (Amminger et al., 2010).
7. Conclusions and Future Directions in Neuroglialpharmacology
For optimal brain function, no class of cells is dispensable. Despite the focus on oligodendrocytes, the goal of this report was to provide a “scaffold” for integrating the largely neuron-centric research efforts with the key roles of glia and especially to the complexity of neuroglial interactions (summarized in section 5 and 6 and Figure 3). In order to help dissipate the historic artificial divide between neurons and glia and encourage an integrated perspective of brain therapeutics, a more inclusive nomenclature (e.g., neuroglialpharmacology) may better serve both clinical and research enterprises. This nomenclature not only enlarges the focus of inquiry to include glia, it also suggests that direct and indirect effects on glia may represent a substantial portion of the efficacy provided by pharmaco- and other therapies (Figure 3).
The hypotheses delineated above are testable through in vivo imaging technologies that provide biomarkers for assessing the trajectory of human myelin development and its subsequent breakdown (Bartzokis et al., 2003; Bartzokis et al., 2004; Hendry et al., 2006; Salat et al., 2005), as well as receptor changes in both gray and white matter (Ding et al., 2004; Pimlott et al., 2004; Vaupel et al., 2005). These technologies, together with genetic as well as clinical and cognitive measures makes it possible to directly test in humans the practical utility of a myelin-focused model of the brain to accelerate medication development. Imaging evidence of promyelination effects of diverse therapeutic interventions in several human psychiatric disorders has recently begun to be measured (Bartzokis et al., 2011a; Bartzokis et al., 2011c; Bartzokis et al., 2009; van der Schot et al., 2009; Venneri and Lane, 2009). Quantifying pharmacologic effects on the brain’s vulnerable oligodendrocyte populations have the potential to elucidate underlying disease processes, mechanisms of action of treatments, and help uncover opportunities for treatment and prevention of both developmental and degenerative brain disorders.
Acknowledgment
This work was supported in part by NIH grants (MH066029; AG027342), Research and Psychiatry Services of the Department of Veterans Affairs, and the RCS Foundation. The author thanks Lori L. Altshuler, M.D., and Keith H. Nuechterlein, Ph.D. for reading the manuscript and providing helpful suggestions.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Author Disclosure - Conflict of Interest George Bartzokis has consulted for and received research funding from Pfizer, Janssen, and Novartis, and consulted for Bristol-Myers Squibb Company.
References
- Aberg ND, Johansson UE, Aberg MA, Hellstrom NA, Lind J, Bull C, Isgaard J, Anderson MF, Oscarsson J, Eriksson PS. Peripheral infusion of insulin-like growth factor-I increases the number of newborn oligodendrocytes in the cerebral cortex of adult hypophysectomized rats. Endocrinology. 2007;148:3765–3772. doi: 10.1210/en.2006-1556. [DOI] [PubMed] [Google Scholar]
- Adamska M, Larroux C, Adamski M, Green K, Lovas E, Koop D, Richards GS, Zwafink C, Degnan BM. Structure and expression of conserved Wnt pathway components in the demosponge Amphimedon queenslandica. Evol Dev. 2011;12:494–518. doi: 10.1111/j.1525-142X.2010.00435.x. [DOI] [PubMed] [Google Scholar]
- Altamura AC, Serati M, Albano A, Paoli RA, Glick ID, Dell'osso B. An epidemiologic and clinical overview of medical and psychopathological comorbidities in major psychoses. Eur Arch Psychiatry Clin Neurosci. 2011;261:489–508. doi: 10.1007/s00406-011-0196-4. [DOI] [PubMed] [Google Scholar]
- Althaus HH, Kloppner S. Mature pig oligodendrocytes rapidly process human recombinant pro-nerve growth factor and do not undergo cell death. J Neurochem. 2006;98:506–517. doi: 10.1111/j.1471-4159.2006.03891.x. [DOI] [PubMed] [Google Scholar]
- Althaus HH, Kloppner S, Klopfleisch S, Schmitz M. Oligodendroglial cells and neurotrophins: a polyphonic cantata in major and minor. J Mol Neurosci. 2008;35:65–79. doi: 10.1007/s12031-008-9053-y. [DOI] [PubMed] [Google Scholar]
- Altshuler LL, Curran JG, Hauser P, Mintz J, Denicoff K, Post R. T2 hyperintensities in bipolar disorder: magnetic resonance imaging comparison and literature meta-analysis. Am J Psychiatry. 1995;152:1139–1144. doi: 10.1176/ajp.152.8.1139. [DOI] [PubMed] [Google Scholar]
- Amminger GP, Schafer MR, Papageorgiou K, Klier CM, Cotton SM, Harrigan SM, Mackinnon A, McGorry PD, Berger GE. Long-chain omega-3 fatty acids for indicated prevention of psychotic disorders: a randomized, placebo-controlled trial. Arch Gen Psychiatry. 2010;67:146–154. doi: 10.1001/archgenpsychiatry.2009.192. [DOI] [PubMed] [Google Scholar]
- Anacker C, Zunszain PA, Cattaneo A, Carvalho LA, Garabedian MJ, Thuret S, Price J, Pariante CM. Antidepressants increase human hippocampal neurogenesis by activating the glucocorticoid receptor. Mol Psychiatry. 2011;16:735–750. doi: 10.1038/mp.2011.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anderson MF, Aberg MA, Nilsson M, Eriksson PS. Insulin-like growth factor-I and neurogenesis in the adult mammalian brain. Brain Res Dev Brain Res. 2002;134:115–122. doi: 10.1016/s0165-3806(02)00277-8. [DOI] [PubMed] [Google Scholar]
- Andreasen NC, Nopoulos P, Magnotta V, Pierson R, Ziebell S, Ho BC. Progressive Brain Change in Schizophrenia: A Prospective Longitudinal Study of First-Episode Schizophrenia. Biol Psychiatry. 2011;70:672–679. doi: 10.1016/j.biopsych.2011.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Astarita G, Jung KM, Berchtold NC, Nguyen VQ, Gillen DL, Head E, Cotman CW, Piomelli D. Deficient liver biosynthesis of docosahexaenoic acid correlates with cognitive impairment in Alzheimer's disease. PLoS One. 2010;5:e12538. doi: 10.1371/journal.pone.0012538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Atanasoski S, Boentert M, De Ventura L, Pohl H, Baranek C, Beier K, Young P, Barbacid M, Suter U. Postnatal Schwann cell proliferation but not myelination is strictly and uniquely dependent on cyclin-dependent kinase 4 (cdk4) Mol Cell Neurosci. 2008;37:519–527. doi: 10.1016/j.mcn.2007.11.005. [DOI] [PubMed] [Google Scholar]
- Azim K, Butt AM. GSK3beta negatively regulates oligodendrocyte differentiation and myelination in vivo. Glia. 2011;59:540–553. doi: 10.1002/glia.21122. [DOI] [PubMed] [Google Scholar]
- Balu DT, Carlson GC, Talbot K, Kazi H, Hill-Smith TE, Easton RM, Birnbaum MJ, Lucki I. Akt1 deficiency in schizophrenia and impairment of hippocampal plasticity and function. Hippocampus. 2010 doi: 10.1002/hipo.20887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barr AM, Fish KN, Markou A. The reelin receptors VLDLR and ApoER2 regulate sensorimotor gating in mice. Neuropharmacology. 2007;52:1114–1123. doi: 10.1016/j.neuropharm.2006.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barradas PC, Vieira RS, De Freitas MS. Selective effect of hypothyroidism on expression of myelin markers during development. J Neurosci Res. 2001;66:254–261. doi: 10.1002/jnr.1218. [DOI] [PubMed] [Google Scholar]
- Bartzokis G. Schizophrenia: breakdown in the well-regulated lifelong process of brain development and maturation. Neuropsychopharmacology. 2002;27:672–683. doi: 10.1016/S0893-133X(02)00364-0. [DOI] [PubMed] [Google Scholar]
- Bartzokis G. Quadratic trajectories of brain myelin content: unifying construct for neuropsychiatric disorders. Neurobiol Aging. 2004;25:49–62. [Google Scholar]
- Bartzokis G. Brain myelination in prevalent neuropsychiatric developmental disorders: Primary and comorbid addiction. Adolescent Psychiatry. 2005;29:55–96. [PMC free article] [PubMed] [Google Scholar]
- Bartzokis G. Acetylcholinesterase inhibitors may improve myelin integrity. Biological Psychiatry. 2007;62:294–301. doi: 10.1016/j.biopsych.2006.08.020. [DOI] [PubMed] [Google Scholar]
- Bartzokis G. Alzheimer's disease as homeostatic responses to age-related myelin breakdown. Neurobiol Aging. 2011a;32:1341–1371. doi: 10.1016/j.neurobiolaging.2009.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartzokis G. Neuroglialpharmacology: white matter pathophysiologies and psychiatric treatments. Frontiers in Bioscience. 2011b;17:2695–2733. doi: 10.2741/3881. [DOI] [PubMed] [Google Scholar]
- Bartzokis G, Altshuler L. Reduced intracortical myelination in schizophrenia. Am J Psychiatry. 2005;162:1229–1230. doi: 10.1176/appi.ajp.162.6.1229-a. [DOI] [PubMed] [Google Scholar]
- Bartzokis G, Beckson M, Lu PH, Edwards N, Bridge P, Mintz J. Brain maturation may be arrested in chronic cocaine addicts. Biol Psychiatry. 2002;51:605–611. doi: 10.1016/s0006-3223(02)01315-x. [DOI] [PubMed] [Google Scholar]
- Bartzokis G, Beckson M, Lu PH, Nuechterlein KH, Edwards N, Mintz J. Age-related changes in frontal and temporal lobe volumes in men: A magnetic resonance imaging study. Arch Gen Psychiatry. 2001;58:461–465. doi: 10.1001/archpsyc.58.5.461. [DOI] [PubMed] [Google Scholar]
- Bartzokis G, Cummings JL, Sultzer D, Henderson VW, Nuechterlein KH, Mintz J. White matter structural integrity in healthy aging adults and patients with Alzheimer disease: a magnetic resonance imaging study. Arch Neurol. 2003;60:393–398. doi: 10.1001/archneur.60.3.393. [DOI] [PubMed] [Google Scholar]
- Bartzokis G, Lu PH, Amar CP, Raven EP, DeTore NR, Altshuler LL, Mintz J, Ventura J, Casaus LR, Luo JS, Subotnik KL, Nuechterlein KH. Long Acting Injection Versus Oral Risperidone in First-Episode Schizophrenia: Differential Impact on White Matter Myelination Trajectory. Schizophrenia Research. 2011a;132:35–41. doi: 10.1016/j.schres.2011.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartzokis G, Lu PH, Raven EP, Amar CP, DeTore NR, Altshuler LL, Mintz J, Ventura J, Casaus LR, Luo JS, Subotnik KL, Nuechterlein KH. Abnormal trajectory of intracortical myelination in schizophrenia implicates white matter in treatment response and outcome. Society of Biological Psychiatry 66th Annual Meeting, San Francisco, CA. Biological Psychiatry. 2011b;69:185S. [Google Scholar]
- Bartzokis G, Lu PH, Raven EP, Amar CP, DeTore NR, Altshuler LL, Mintz J, Ventura J, Casaus LR, Luo JS, Subotnik KL, Nuechterlein KH. Abnormal Trajectory of Intracortical Myelination in Schizophrenia Implicates White Matter in Treatment Response and Outcomes. 2011c (Submitted) [Google Scholar]
- Bartzokis G, Lu PH, Stewart SB, Oluwadara B, Lucas AJ, Pantages J, Pratt E, Sherin JE, Altshuler LL, Mintz J, Gitlin MJ, Subotnik KL, Nuechterlein KH. In vivo evidence of differential impact of typical and atypical antipsychotics on intracortical myelin in adults with schizophrenia. Schizophr Res. 2009;113:322–331. doi: 10.1016/j.schres.2009.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartzokis G, Lu PH, Tingus K, Mendez MF, Richard A, Peters DG, Oluwadara B, Barrall KA, Finn JP, Villablanca P, Thompson PM, Mintz J. Lifespan trajectory of myelin integrity and maximum motor speed. Neurobiol Aging. 2010;31:1554–1562. doi: 10.1016/j.neurobiolaging.2008.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartzokis G, Lu PH, Tingus K, Peters DG, Amar CP, Tishler TA, Finn JP, Villablanca P, Altshuler LL, Mintz J, Neely E, Connor JR. Gender and iron genes may modify associations between brain iron and memory in healthy aging. Neuropsychopharmacology. 2011d;36:1375–1384. doi: 10.1038/npp.2011.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartzokis G, Sultzer D, Lu PH, Nuechterlein KH, Mintz J, Cummings J. Heterogeneous agerelated breakdown of white matter structural integrity: Implications for cortical "disconnection" in aging and Alzheimer's disease. Neurobiol Aging. 2004;25:843–851. doi: 10.1016/j.neurobiolaging.2003.09.005. [DOI] [PubMed] [Google Scholar]
- Bauer M, Goetz T, Glenn T, Whybrow PC. The thyroid-brain interaction in thyroid disorders and mood disorders. J Neuroendocrinol. 2008;20:1101–1114. doi: 10.1111/j.1365-2826.2008.01774.x. [DOI] [PubMed] [Google Scholar]
- Bauer M, London ED, Rasgon N, Berman SM, Frye MA, Altshuler LL, Mandelkern MA, Bramen J, Voytek B, Woods R, Mazziotta JC, Whybrow PC. Supraphysiological doses of levothyroxine alter regional cerebral metabolism and improve mood in bipolar depression. Mol Psychiatry. 2005;10:456–469. doi: 10.1038/sj.mp.4001647. [DOI] [PubMed] [Google Scholar]
- Beaulieu JM, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63:182–217. doi: 10.1124/pr.110.002642. [DOI] [PubMed] [Google Scholar]
- Beaulieu JM, Sotnikova TD, Marion S, Lefkowitz RJ, Gainetdinov RR, Caron MG. An Akt/beta-arrestin 2/PP2A signaling complex mediates dopaminergic neurotransmission and behavior. Cell. 2005;122:261–273. doi: 10.1016/j.cell.2005.05.012. [DOI] [PubMed] [Google Scholar]
- Beaulieu JM, Sotnikova TD, Yao WD, Kockeritz L, Woodgett JR, Gainetdinov RR, Caron MG. Lithium antagonizes dopamine-dependent behaviors mediated by an AKT/glycogen synthase kinase 3 signaling cascade. Proc Natl Acad Sci U S A. 2004;101:5099–5104. doi: 10.1073/pnas.0307921101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beffert U, Weeber EJ, Durudas A, Qiu S, Masiulis I, Sweatt JD, Li WP, Adelmann G, Frotscher M, Hammer RE, Herz J. Modulation of synaptic plasticity and memory by reelin involves differential splicing of the lipoprotein receptor apoer2. Neuron. 2005;47:567–579. doi: 10.1016/j.neuron.2005.07.007. [DOI] [PubMed] [Google Scholar]
- Belachew S, Rogister B, Rigo JM, Malgrange B, Moonen G. Neurotransmitter-mediated regulation of CNS myelination: a review. Acta Neurol Belg. 1999;99:21–31. [PubMed] [Google Scholar]
- Bennett GW, Ballard TM, Watson CD, Fone KC. Effect of neuropeptides on cognitive function. Exp Gerontol. 1997;32:451–469. doi: 10.1016/s0531-5565(96)00159-3. [DOI] [PubMed] [Google Scholar]
- Berretta S. Extracellular matrix abnormalities in schizophrenia. Neuropharmacology. 2011 doi: 10.1016/j.neuropharm.2011.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bibollet-Bahena O, Almazan G. IGF-1-stimulated protein synthesis in oligodendrocyte progenitors requires PI3K/mTOR/Akt and MEK/ERK pathways. J Neurochem. 2009;109:1440–1451. doi: 10.1111/j.1471-4159.2009.06071.x. [DOI] [PubMed] [Google Scholar]
- Bitner RS, Bunnelle WH, Decker MW, Drescher KU, Kohlhaas KL, Markosyan S, Marsh KC, Nikkel AL, Browman K, Radek R, Anderson DJ, Buccafusco J, Gopalakrishnan M. In vivo pharmacological characterization of a novel selective alpha7 neuronal nicotinic acetylcholine receptor agonist ABT-107: preclinical considerations in Alzheimer's disease. J Pharmacol Exp Ther. 2010;334:875–886. doi: 10.1124/jpet.110.167213. [DOI] [PubMed] [Google Scholar]
- Blasi G, Napolitano F, Ursini G, Taurisano P, Romano R, Caforio G, Fazio L, Gelao B, Di Giorgio A, Iacovelli L, Sinibaldi L, Popolizio T, Usiello A, Bertolino A. DRD2/AKT1 interaction on D2 c-AMP independent signaling, attentional processing, and response to olanzapine treatment in schizophrenia. Proc Natl Acad Sci U S A. 2011;108:1158–1163. doi: 10.1073/pnas.1013535108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bolos M, Fernandez S, Torres-Aleman I. Oral administration of a GSK3 inhibitor increases brain insulin-like growth factor I levels. J Biol Chem. 2011;285:17693–17700. doi: 10.1074/jbc.M109.096594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bondy CA, Cheng CM. Signaling by insulin-like growth factor 1 in brain. Eur J Pharmacol. 2004;490:25–31. doi: 10.1016/j.ejphar.2004.02.042. [DOI] [PubMed] [Google Scholar]
- Botella-Lopez A, Cuchillo-Ibanez I, Cotrufo T, Mok SS, Li QX, Barquero MS, Dierssen M, Soriano E, Saez-Valero J. Beta-amyloid controls altered Reelin expression and processing in Alzheimer's disease. Neurobiol Dis. 2010;37:682–691. doi: 10.1016/j.nbd.2009.12.006. [DOI] [PubMed] [Google Scholar]
- Bourque M, Liu B, Dluzen DE, Di Paolo T. Sex differences in methamphetamine toxicity in mice: Effect on brain dopamine signaling pathways. Psychoneuroendocrinology. 2011;36:955–969. doi: 10.1016/j.psyneuen.2010.12.007. [DOI] [PubMed] [Google Scholar]
- Brambilla P, Bellani M, Yeh PH, Soares JC. Myelination in bipolar patients and the effects of mood stabilizers on brain anatomy. Curr Pharm Des. 2009;15:2632–2636. doi: 10.2174/138161209788957519. [DOI] [PubMed] [Google Scholar]
- Brosda J, Dietz F, Koch M. Impairment of cognitive performance after reelin knockdown in the medial prefrontal cortex of pubertal or adult rats. Neurobiol Dis. 2011;44:239–247. doi: 10.1016/j.nbd.2011.07.008. [DOI] [PubMed] [Google Scholar]
- Burdick KE, Goldberg TE, Cornblatt BA, Keefe RS, Gopin CB, Derosse P, Braga RJ, Malhotra AK. The MATRICS Consensus Cognitive Battery in Patients with Bipolar I Disorder. Neuropsychopharmacology. 2011;36:1587–1592. doi: 10.1038/npp.2011.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butt AM. Neurotransmitter-mediated calcium signalling in oligodendrocyte physiology and pathology. Glia. 2006;54:666–675. doi: 10.1002/glia.20424. [DOI] [PubMed] [Google Scholar]
- Butt AM. ATP: A ubiquitous gliotransmitter integrating neuron-glial networks. Semin Cell Dev Biol. 2011;22:205–213. doi: 10.1016/j.semcdb.2011.02.023. [DOI] [PubMed] [Google Scholar]
- Butt AM, Berry M. Oligodendrocytes and the control of myelination in vivo: new insights from the rat anterior medullary velum. J.Neurosci. Res. 2000;59:477–488. doi: 10.1002/(SICI)1097-4547(20000215)59:4<477::AID-JNR2>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
- Buzsaki G, Draguhn A. Neuronal oscillations in cortical networks. Science. 2004;304:1926–1929. doi: 10.1126/science.1099745. [DOI] [PubMed] [Google Scholar]
- Caccamo A, Majumder S, Richardson A, Strong R, Oddo S. Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: effects on cognitive impairments. J Biol Chem. 2010;285:13107–13120. doi: 10.1074/jbc.M110.100420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caillava C, Vandenbosch R, Jablonska B, Deboux C, Spigoni G, Gallo V, Malgrange B, Baron-Van Evercooren A. Cdk2 loss accelerates precursor differentiation and remyelination in the adult central nervous system. J Cell Biol. 2011;193:397–407. doi: 10.1083/jcb.201004146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calabrese M, Filippi M, Rovaris M, Bernardi V, Atzori M, Mattisi I, Favaretto A, Grossi P, Barachino L, Rinaldi L, Romualdi C, Perini P, Gallo P. Evidence for relative cortical sparing in benign multiple sclerosis: a longitudinal magnetic resonance imaging study. Mult Scler. 2009;15:36–41. doi: 10.1177/1352458508096686. [DOI] [PubMed] [Google Scholar]
- Cao X, Kambe F, Yamauchi M, Seo H. Thyroid-hormone-dependent activation of the phosphoinositide 3-kinase/Akt cascade requires Src and enhances neuronal survival. Biochem J. 2009;424:201–209. doi: 10.1042/BJ20090643. [DOI] [PubMed] [Google Scholar]
- Carson MJ, Behringer RR, Brinster RL, McMorris FA. Insulin-like growth factor I increases brain growth and central nervous system myelination in transgenic mice. Neuron. 1993;10:729–740. doi: 10.1016/0896-6273(93)90173-o. [DOI] [PubMed] [Google Scholar]
- Chan A, Shea TB. Folate deprivation increases presenilin expression, gamma-secretase activity, and Abeta levels in murine brain: potentiation by ApoE deficiency and alleviation by dietary S-adenosyl methionine. J Neurochem. 2007;102:753–760. doi: 10.1111/j.1471-4159.2007.04589.x. [DOI] [PubMed] [Google Scholar]
- Clarke R, Smith AD, Jobst KA, Refsum H, Sutton L, Ueland PM. Folate, vitamin B12, and serum total homocysteine levels in confirmed Alzheimer disease [see comments] Arch.Neurol. 1998;55:1449–1455. doi: 10.1001/archneur.55.11.1449. [DOI] [PubMed] [Google Scholar]
- Coelho RP, Yuelling LM, Fuss B, Sato-Bigbee C. Neurotrophin-3 targets the translational initiation machinery in oligodendrocytes. Glia. 2009;57:1754–1764. doi: 10.1002/glia.20888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corfas G, Roy K, Buxbaum JD. Neuregulin 1-erbB signaling and the molecular/cellular basis of schizophrenia. Nat Neurosci. 2004;7:575–580. doi: 10.1038/nn1258. [DOI] [PubMed] [Google Scholar]
- Cui QL, Fogle E, Almazan G. Muscarinic acetylcholine receptors mediate oligodendrocyte progenitor survival through Src-like tyrosine kinases and PI3K/Akt pathways. Neurochem Int. 2006;48:383–393. doi: 10.1016/j.neuint.2005.11.014. [DOI] [PubMed] [Google Scholar]
- D'Ercole AJ, Ye P. Expanding The Mind: IGF-I and Brain Development. Endocrinology. 2008;149:5958–5962. doi: 10.1210/en.2008-0920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis KL, Stewart DG, Friedman JI, Buchsbaum M, Harvey PD, Hof PR, Buxbaum J, Haroutunian V. White matter changes in schizophrenia: evidence for myelin-related dysfunction. Arch Gen Psychiatry. 2003;60:443–456. doi: 10.1001/archpsyc.60.5.443. [DOI] [PubMed] [Google Scholar]
- Day JJ, Sweatt JD. Epigenetic mechanisms in cognition. Neuron. 2011;70:813–829. doi: 10.1016/j.neuron.2011.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Lau LM, Smith AD, Refsum H, Johnston C, Breteler MM. Plasma vitamin B12 status and cerebral white-matter lesions. J Neurol Neurosurg Psychiatry. 2009;80:149–157. doi: 10.1136/jnnp.2008.149286. [DOI] [PubMed] [Google Scholar]
- De Sarno P, Axtell RC, Raman C, Roth KA, Alessi DR, Jope RS. Lithium prevents and ameliorates experimental autoimmune encephalomyelitis. J Immunol. 2008;181:338–345. doi: 10.4049/jimmunol.181.1.338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Sarno P, Bijur GN, Zmijewska AA, Li X, Jope RS. In vivo regulation of GSK3 phosphorylation by cholinergic and NMDA receptors. Neurobiol Aging. 2006;27:413–422. doi: 10.1016/j.neurobiolaging.2005.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Descarries L, Watkins KC, Garcia S, Bosler O, Doucet G. Dual character, asynaptic and synaptic, of the dopamine innervation in adult rat neostriatum: a quantitative autoradiographic and immunocytochemical analysis. J Comp Neurol. 1996;375:167–186. doi: 10.1002/(SICI)1096-9861(19961111)375:2<167::AID-CNE1>3.0.CO;2-0. [DOI] [PubMed] [Google Scholar]
- Ding YS, Fowler JS, Logan J, Wang GJ, Telang F, Garza V, Biegon A, Pareto D, Rooney W, Shea C, Alexoff D, Volkow ND, Vocci F. 6-[18F]Fluoro-A-85380, a new PET tracer for the nicotinic acetylcholine receptor: studies in the human brain and in vivo demonstration of specific binding in white matter. Synapse. 2004;53:184–189. doi: 10.1002/syn.20051. [DOI] [PubMed] [Google Scholar]
- Du Y, Lercher LD, Zhou R, Dreyfus CF. Mitogen-activated protein kinase pathway mediates effects of brain-derived neurotrophic factor on differentiation of basal forebrain oligodendrocytes. J Neurosci Res. 2006;84:1692–1702. doi: 10.1002/jnr.21080. [DOI] [PubMed] [Google Scholar]
- Duman RS, Li N, Liu RJ, Duric V, Aghajanian G. Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology. 2012;62:35–41. doi: 10.1016/j.neuropharm.2011.08.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dwork AJ, Mancevski B, Rosoklija G. White matter and cognitive function in schizophrenia. Int J Neuropsychopharmacol. 2007:1–24. doi: 10.1017/S1461145707007638. [DOI] [PubMed] [Google Scholar]
- Emamian ES, Hall D, Birnbaum MJ, Karayiorgou M, Gogos JA. Convergent evidence for impaired AKT1-GSK3beta signaling in schizophrenia. Nat Genet. 2004;36:131–137. doi: 10.1038/ng1296. [DOI] [PubMed] [Google Scholar]
- Emsley R, Rabinowitz J, Medori R. Remission in early psychosis: Rates, predictors, and clinical and functional outcome correlates. Schizophr Res. 2007;89:129–139. doi: 10.1016/j.schres.2006.09.013. [DOI] [PubMed] [Google Scholar]
- Engmann O, Giese KP. Crosstalk between Cdk5 and GSK3beta: Implications for Alzheimer's Disease. Front Mol Neurosci. 2009;2:2. doi: 10.3389/neuro.02.002.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Engmann O, Hortobagyi T, Pidsley R, Troakes C, Bernstein HG, Kreutz MR, Mill J, Nikolic M, Giese KP. Schizophrenia is associated with dysregulation of a Cdk5 activator that regulates synaptic protein expression and cognition. Brain. 2011;134:2408–2421. doi: 10.1093/brain/awr155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Etxeberria A, Mangin JM, Aguirre A, Gallo V. Adult-born SVZ progenitors receive transient synapses during remyelination in corpus callosum. Nat Neurosci. 2010;13:287–289. doi: 10.1038/nn.2500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fatemi SH, Reutiman TJ, Folsom TD. Chronic psychotropic drug treatment causes differential expression of Reelin signaling system in frontal cortex of rats. Schizophr Res. 2009;111:138–152. doi: 10.1016/j.schres.2009.03.002. [DOI] [PubMed] [Google Scholar]
- Fatemi SH, Snow AV, Stary JM, Araghi-Niknam M, Reutiman TJ, Lee S, Brooks AI, Pearce DA. Reelin signaling is impaired in autism. Biol Psychiatry. 2005a;57:777–787. doi: 10.1016/j.biopsych.2004.12.018. [DOI] [PubMed] [Google Scholar]
- Fatemi SH, Stary JM, Earle JA, Araghi-Niknam M, Eagan E. GABAergic dysfunction in schizophrenia and mood disorders as reflected by decreased levels of glutamic acid decarboxylase 65 and 67 kDa and Reelin proteins in cerebellum. Schizophr Res. 2005b;72:109–122. doi: 10.1016/j.schres.2004.02.017. [DOI] [PubMed] [Google Scholar]
- Fei W, Aixi Y, Danmou X, Wusheng K, Zhengren P, Ting R. The mood stabilizer valproic acid induces proliferation and myelination of rat Schwann cells. Neurosci Res. 2011;70:383–390. doi: 10.1016/j.neures.2011.04.002. [DOI] [PubMed] [Google Scholar]
- Fields RD, Burnstock G. Purinergic signalling in neuron-glia interactions. Nat Rev Neurosci. 2006;7:423–436. doi: 10.1038/nrn1928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flores AI, Narayanan SP, Morse EN, Shick HE, Yin X, Kidd G, Avila RL, Kirschner DA, Macklin WB. Constitutively active Akt induces enhanced myelination in the CNS. J Neurosci. 2008;28:7174–7183. doi: 10.1523/JNEUROSCI.0150-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fragoso G, Haines JD, Roberston J, Pedraza L, Mushynski WE, Almazan G. p38 mitogen-activated protein kinase is required for central nervous system myelination. Glia. 2007;55:1531–1541. doi: 10.1002/glia.20567. [DOI] [PubMed] [Google Scholar]
- Frederick TJ, Min J, Altieri SC, Mitchell NE, Wood TL. Synergistic induction of cyclin D1 in oligodendrocyte progenitor cells by IGF-I and FGF-2 requires differential stimulation of multiple signaling pathways. Glia. 2007;55:1011–1022. doi: 10.1002/glia.20520. [DOI] [PubMed] [Google Scholar]
- Frederick TJ, Wood TL. IGF-I and FGF-2 coordinately enhance cyclin D1 and cyclin E-cdk2 association and activity to promote G1 progression in oligodendrocyte progenitor cells. Mol Cell Neurosci. 2004;25:480–492. doi: 10.1016/j.mcn.2003.11.015. [DOI] [PubMed] [Google Scholar]
- Freude S, Leeser U, Muller M, Hettich MM, Udelhoven M, Schilbach K, Tobe K, Kadowaki T, Kohler C, Schroder H, Krone W, Bruning JC, Schubert M. IRS-2 branch of IGF-1 receptor signaling is essential for appropriate timing of myelination. J Neurochem. 2008;107:907–917. doi: 10.1111/j.1471-4159.2008.05631.x. [DOI] [PubMed] [Google Scholar]
- Friedman JI, Tang C, Carpenter D, Buchsbaum M, Schmeidler J, Flanagan L, Golembo S, Kanellopoulou I, Ng J, Hof PR, Harvey PD, Tsopelas ND, Stewart D, Davis KL. Diffusion tensor imaging findings in first-episode and chronic schizophrenia patients. Am J Psychiatry. 2008;165:1024–1032. doi: 10.1176/appi.ajp.2008.07101640. [DOI] [PubMed] [Google Scholar]
- Fuxe K, Agnati LF. In: Transmission in the Brain. Fuxe K, Agnati LF, editors. New York: Raven; 1991. pp. 1–9. [Google Scholar]
- Fyffe-Maricich SL, Karlo JC, Landreth GE, Miller RH. The ERK2 mitogen-activated protein kinase regulates the timing of oligodendrocyte differentiation. J Neurosci. 2011;31:843–850. doi: 10.1523/JNEUROSCI.3239-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garelick MG, Kennedy BK. TOR on the brain. Exp Gerontol. 2011;46:155–163. doi: 10.1016/j.exger.2010.08.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goebbels S, Oltrogge JH, Kemper R, Heilmann I, Bormuth I, Wolfer S, Wichert SP, Mobius W, Liu X, Lappe-Siefke C, Rossner MJ, Groszer M, Suter U, Frahm J, Boretius S, Nave KA. Elevated phosphatidylinositol 3,4,5-trisphosphate in glia triggers cell-autonomous membrane wrapping and myelination. J Neurosci. 2010;30:8953–8964. doi: 10.1523/JNEUROSCI.0219-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gomez JM. Growth hormone and insulin-like growth factor-I as an endocrine axis in Alzheimer's disease. Endocr Metab Immune Disord Drug Targets. 2008;8:143–151. doi: 10.2174/187153008784534367. [DOI] [PubMed] [Google Scholar]
- Graber JJ, Sherman FT, Kaufmann H, Kolodny EH, Sathe S. Vitamin B12-responsive severe leukoencephalopathy and autonomic dysfunction in a patient with "normal" serum B12 levels. J Neurol Neurosurg Psychiatry. 2011;81:1369–1371. doi: 10.1136/jnnp.2009.178657. [DOI] [PubMed] [Google Scholar]
- Greco SJ, Sarkar S, Casadesus G, Zhu X, Smith MA, Ashford JW, Johnston JM, Tezapsidis N. Leptin inhibits glycogen synthase kinase-3 beta to prevent tau phosphorylation in neuronal cells. Neurosci Lett. 2009;455:191–194. doi: 10.1016/j.neulet.2009.03.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gudz TI, Komuro H, Macklin WB. Glutamate stimulates oligodendrocyte progenitor migration mediated via an alphav integrin/myelin proteolipid protein complex. J Neurosci. 2006;26:2458–2466. doi: 10.1523/JNEUROSCI.4054-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guidotti A, Auta J, Chen Y, Davis JM, Dong E, Gavin DP, Grayson DR, Matrisciano F, Pinna G, Satta R, Sharma RP, Tremolizzo L, Tueting P. Epigenetic GABAergic targets in schizophrenia and bipolar disorder. Neuropharmacology. 2011;60:1007–1016. doi: 10.1016/j.neuropharm.2010.10.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haines JD, Fang J, Mushynski WE, Almazan G. Mitogen-activated protein kinase activated protein kinase 2 (MK2) participates in p38 MAPK regulated control of oligodendrocyte differentiation. Glia. 2010;58:1384–1393. doi: 10.1002/glia.21014. [DOI] [PubMed] [Google Scholar]
- Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, Pahor M, Javors MA, Fernandez E, Miller RA. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–395. doi: 10.1038/nature08221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harsan LA, Steibel J, Zaremba A, Agin A, Sapin R, Poulet P, Guignard B, Parizel N, Grucker D, Boehm N, Miller RH, Ghandour MS. Recovery from chronic demyelination by thyroid hormone therapy: myelinogenesis induction and assessment by diffusion tensor magnetic resonance imaging. J Neurosci. 2008;28:14189–14201. doi: 10.1523/JNEUROSCI.4453-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hartman BK, Agrawal HC, Agrawal D, Kalmbach S. Development and maturation of central nervous system myelin: comparison of immunohistochemical localization of proteolipid protein and basic protein in myelin and oligodendrocytes. Proc Natl Acad Sci U S A. 1982;79:4217–4220. doi: 10.1073/pnas.79.13.4217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hendry J, Devito T, Gelman N, Densmore M, Rajakumar N, Pavlosky W, Williamson PC, Thompson PM, Drost DJ, Nicolson R. White matter abnormalities in autism detected through transverse relaxation time imaging. Neuroimage. 2006;29:1049–1057. doi: 10.1016/j.neuroimage.2005.08.039. [DOI] [PubMed] [Google Scholar]
- Ho BC, Andreasen NC, Ziebell S, Pierson R, Magnotta V. Long-term antipsychotic treatment and brain volumes: a longitudinal study of first-episode schizophrenia. Arch Gen Psychiatry. 2011;68:128–137. doi: 10.1001/archgenpsychiatry.2010.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hong LE, Summerfelt A, Mitchell BD, McMahon RP, Wonodi I, Buchanan RW, Thaker GK. Sensory gating endophenotype based on its neural oscillatory pattern and heritability estimate. Arch Gen Psychiatry. 2008;65:1008–1016. doi: 10.1001/archpsyc.65.9.1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howes OD, Bose SK, Turkheimer F, Valli I, Egerton A, Valmaggia LR, Murray RM, McGuire P. Dopamine Synthesis Capacity Before Onset of Psychosis: A Prospective [18F]-DOPA PET Imaging Study. Am J Psychiatry [Epub ahead of print] 2011 doi: 10.1176/appi.ajp.2011.11010160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsu LS, Liang CJ, Tseng CY, Yeh CW, Tsai JN. Zebrafish Cyclin-Dependent Protein Kinase-Like 1 (zcdkl1): Identification and Functional Characterization. Int J Mol Sci. 2011;12:3606–3617. doi: 10.3390/ijms12063606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Impagnatiello F, Guidotti AR, Pesold C, Dwivedi Y, Caruncho H, Pisu MG, Uzunov DP, Smalheiser NR, Davis JM, Pandey GN, Pappas GD, Tueting P, Sharma RP, Costa E. A decrease of reelin expression as a putative vulnerability factor in schizophrenia. Proc Natl Acad Sci U S A. 1998;95:15718–15723. doi: 10.1073/pnas.95.26.15718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jagannathan K, Calhoun VD, Gelernter J, Stevens MC, Liu J, Bolognani F, Windemuth A, Ruano G, Assaf M, Pearlson GD. Genetic Associations of Brain Structural Networks in Schizophrenia: A Preliminary Study. Biol Psychiatry. 2010;68:657–666. doi: 10.1016/j.biopsych.2010.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jope RS, Johnson GV. The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci. 2004;29:95–102. doi: 10.1016/j.tibs.2003.12.004. [DOI] [PubMed] [Google Scholar]
- Kaes T. Die Grosshirnrinde des Menschen in ihren Massen und in ihrem Fasergehalt. Jena: Gustav Fisher; 1907. [Google Scholar]
- Kang UG, Roh MS, Jung JR, Shin SY, Lee YH, Park JB, Kim YS. Activation of protein kinase B (Akt) signaling after electroconvulsive shock in the rat hippocampus. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28:41–44. doi: 10.1016/S0278-5846(03)00173-8. [DOI] [PubMed] [Google Scholar]
- Karadottir R, Attwell D. Neurotransmitter receptors in the life and death of oligodendrocytes. Neuroscience. 2007;145:1426–1438. doi: 10.1016/j.neuroscience.2006.08.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karege F, Perroud N, Burkhardt S, Fernandez R, Ballmann E, La Harpe R, Malafosse A. Alterations in phosphatidylinositol 3-kinase activity and PTEN phosphatase in the prefrontal cortex of depressed suicide victims. Neuropsychobiology. 2011;63:224–231. doi: 10.1159/000322145. [DOI] [PubMed] [Google Scholar]
- Karege F, Perroud N, Burkhardt S, Schwald M, Ballmann E, La Harpe R, Malafosse A. Alteration in kinase activity but not in protein levels of protein kinase B and glycogen synthase kinase-3beta in ventral prefrontal cortex of depressed suicide victims. Biol Psychiatry. 2007;61:240–245. doi: 10.1016/j.biopsych.2006.04.036. [DOI] [PubMed] [Google Scholar]
- Karege F, Perroud N, Schurhoff F, Meary A, Marillier G, Burkhardt S, Ballmann E, Fernandez R, Jamain S, Leboyer M, La Harpe R, Malafosse A. Association of AKT1 gene variants and protein expression in both schizophrenia and bipolar disorder. Genes Brain Behav. 2010;9:503–511. doi: 10.1111/j.1601-183X.2010.00578.x. [DOI] [PubMed] [Google Scholar]
- Keith S. Use of long-acting risperidone in psychiatric disorders: focus on efficacy, safety and costeffectiveness. Expert Rev Neurother. 2009;9:9–31. doi: 10.1586/14737175.9.1.9. [DOI] [PubMed] [Google Scholar]
- Kemper T. Neuroanatomical and neuropathological changes during aging and dementia. In: Albert M, Knoefel J, editors. Clinical Neurology of Aging. New York: Oxford University Press; 1994. pp. 3–67. [Google Scholar]
- Kempton MJ, Geddes JR, Ettinger U, Williams SC, Grasby PM. Meta-analysis, database, and meta-regression of 98 structural imaging studies in bipolar disorder. Arch Gen Psychiatry. 2008;65:1017–1032. doi: 10.1001/archpsyc.65.9.1017. [DOI] [PubMed] [Google Scholar]
- Kerns D, Vong GS, Barley K, Dracheva S, Katsel P, Casaccia P, Haroutunian V, Byne W. Gene expression abnormalities and oligodendrocyte deficits in the internal capsule in schizophrenia. Schizophr Res. 2010;120:150–158. doi: 10.1016/j.schres.2010.04.012. [DOI] [PubMed] [Google Scholar]
- Kessaris N, Fogarty M, Iannarelli P, Grist M, Wegner M, Richardson WD. Competing waves of oligodendrocytes in the forebrain and postnatal elimination of an embryonic lineage. Nat Neurosci. 2006;9:173–179. doi: 10.1038/nn1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim AJ, Shi Y, Austin RC, Werstuck GH. Valproate protects cells from ER stress-induced lipid accumulation and apoptosis by inhibiting glycogen synthase kinase-3. J Cell Sci. 2005;118:89–99. doi: 10.1242/jcs.01562. [DOI] [PubMed] [Google Scholar]
- Kim JM, Stewart R, Kim SW, Shin IS, Yang SJ, Shin HY, Yoon JS. Changes in folate, vitamin B12 and homocysteine associated with incident dementia. J Neurol Neurosurg Psychiatry. 2008;79:864–868. doi: 10.1136/jnnp.2007.131482. [DOI] [PubMed] [Google Scholar]
- Kimura F, Itami C. Myelination and isochronicity in neural networks. Front Neuroanat. 2009;3:12. doi: 10.3389/neuro.05.012.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klausberger T, Somogyi P. Neuronal diversity and temporal dynamics: the unity of hippocampal circuit operations. Science. 2008;321:53–57. doi: 10.1126/science.1149381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kocherhans S, Madhusudan A, Doehner J, Breu KS, Nitsch RM, Fritschy JM, Knuesel I. Reduced Reelin expression accelerates amyloid-beta plaque formation and tau pathology in transgenic Alzheimer's disease mice. J Neurosci. 2010;30:9228–9240. doi: 10.1523/JNEUROSCI.0418-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koenen KC, Moffitt TE, Roberts AL, Martin LT, Kubzansky L, Harrington H, Poulton R, Caspi A. Childhood IQ and adult mental disorders: a test of the cognitive reserve hypothesis. Am J Psychiatry. 2009;166:50–57. doi: 10.1176/appi.ajp.2008.08030343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kolodziejczyk K, Saab AS, Nave KA, Attwell D. Why do oligodendrocyte lineage cells express glutamate receptors? F1000 Biol Rep. 2010;2:57. doi: 10.3410/B2-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Konopaske GT, Dorph-Petersen KA, Sweet RA, Pierri JN, Zhang W, Sampson AR, Lewis DA. Effect of chronic antipsychotic exposure on astrocyte and oligodendrocyte numbers in macaque monkeys. Biol Psychiatry. 2008;63:759–765. doi: 10.1016/j.biopsych.2007.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kramer PL, Xu H, Woltjer RL, Westaway SK, Clark D, Erten-Lyons D, Kaye JA, Welsh-Bohmer KA, Troncoso JC, Markesbery WR, Petersen RC, Turner RS, Kukull WA, Bennett DA, Galasko D, Morris JC, Ott J. Alzheimer disease pathology in cognitively healthy elderly: A genome-wide study. Neurobiol Aging. 2011;32:2113–2122. doi: 10.1016/j.neurobiolaging.2010.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S, Kahn MA, Dinh L, de Vellis J. NT-3-mediated TrkC receptor activation promotes proliferation and cell survival of rodent progenitor oligodendrocyte cells in vitro and in vivo. J Neurosci Res. 1998;54:754–765. doi: 10.1002/(SICI)1097-4547(19981215)54:6<754::AID-JNR3>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
- Lai WS, Xu B, Westphal KG, Paterlini M, Olivier B, Pavlidis P, Karayiorgou M, Gogos JA. Akt1 deficiency affects neuronal morphology and predisposes to abnormalities in prefrontal cortex functioning. Proc Natl Acad Sci U S A. 2006;103:16906–16911. doi: 10.1073/pnas.0604994103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larocque D, Galarneau A, Liu HN, Scott M, Almazan G, Richard S. Protection of p27(Kip1) mRNA by quaking RNA binding proteins promotes oligodendrocyte differentiation. Nat Neurosci. 2005;8:27–33. doi: 10.1038/nn1359. [DOI] [PubMed] [Google Scholar]
- Lebel M, Patenaude C, Allyson J, Massicotte G, Cyr M. Dopamine D1 receptor activation induces tau phosphorylation via cdk5 and GSK3 signaling pathways. Neuropharmacology. 2009;57:392–402. doi: 10.1016/j.neuropharm.2009.06.041. [DOI] [PubMed] [Google Scholar]
- Lesuisse D, Dutruc-Rosset G, Tiraboschi G, Dreyer MK, Maignan S, Chevalier A, Halley F, Bertrand P, Burgevin MC, Quarteronet D, Rooney T. Rational design of potent GSK3beta inhibitors with selectivity for Cdk1 and Cdk2. Bioorg Med Chem Lett. 2010;20:1985–1989. doi: 10.1016/j.bmcl.2010.01.114. [DOI] [PubMed] [Google Scholar]
- Levine JM, Reynolds R, Fawcett JW. The oligodendrocyte precursor cell in health and disease. Trends Neurosci. 2001;24:39–47. doi: 10.1016/s0166-2236(00)01691-x. [DOI] [PubMed] [Google Scholar]
- Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, Li XY, Aghajanian G, Duman RS. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010a;329:959–964. doi: 10.1126/science.1190287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Jope RS. Is glycogen synthase kinase-3 a central modulator in mood regulation? Neuropsychopharmacology. 2010;35:2143–2154. doi: 10.1038/npp.2010.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Liu M, Cai Z, Wang G, Li X. Regulation of glycogen synthase kinase-3 during bipolar mania treatment. Bipolar Disord. 2010b;12:741–752. doi: 10.1111/j.1399-5618.2010.00866.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Rosborough KM, Friedman AB, Zhu W, Roth KA. Regulation of mouse brain glycogen synthase kinase-3 by atypical antipsychotics. Int J Neuropsychopharmacol. 2007;10:7–19. doi: 10.1017/S1461145706006547. [DOI] [PubMed] [Google Scholar]
- Lieberman J, Jody D, Geisler S, Alvir J, Loebel A, Szymanski S, Woerner M, Borenstein M. Time course and biologic correlates of treatment response in first-episode schizophrenia. Arch Gen Psychiatry. 1993;50:369–376. doi: 10.1001/archpsyc.1993.01820170047006. [DOI] [PubMed] [Google Scholar]
- Linkenkaer-Hansen K, Smit DJ, Barkil A, van Beijsterveldt TE, Brussaard AB, Boomsma DI, van Ooyen A, de Geus EJ. Genetic contributions to long-range temporal correlations in ongoing oscillations. J Neurosci. 2007;27:13882–13889. doi: 10.1523/JNEUROSCI.3083-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Llorens-Martin M, Torres-Aleman I, Trejo JL. Exercise modulates insulin-like growth factor 1- dependent and -independent effects on adult hippocampal neurogenesis and behaviour. Mol Cell Neurosci. 2010;44:109–117. doi: 10.1016/j.mcn.2010.02.006. [DOI] [PubMed] [Google Scholar]
- Luo HR, Hattori H, Hossain MA, Hester L, Huang Y, Lee-Kwon W, Donowitz M, Nagata E, Snyder SH. Akt as a mediator of cell death. Proc Natl Acad Sci U S A. 2003;100:11712–11717. doi: 10.1073/pnas.1634990100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo L, Stopa EG. Thyrotropin releasing hormone inhibits tau phosphorylation by dual signaling pathways in hippocampal neurons. J Alzheimers Dis. 2004;6:527–536. doi: 10.3233/jad-2004-6510. [DOI] [PubMed] [Google Scholar]
- Luo L, Yano N, Mao Q, Jackson IM, Stopa EG. Thyrotropin releasing hormone (TRH) in the hippocampus of Alzheimer patients. J Alzheimers Dis. 2002;4:97–103. doi: 10.3233/jad-2002-4204. [DOI] [PubMed] [Google Scholar]
- Ma T, Hoeffer CA, Capetillo-Zarate E, Yu F, Wong H, Lin MT, Tampellini D, Klann E, Blitzer RD, Gouras GK. Dysregulation of the mTOR pathway mediates impairment of synaptic plasticity in a mouse model of Alzheimer's disease. PLoS One. 2010;5:e12845. doi: 10.1371/journal.pone.0012845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mangin JM, Gallo V. The curious case of NG2 cells: transient trend or game changer? ASN Neuro. 2011;3:e00052. doi: 10.1042/AN20110001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marner L, Nyengaard JR, Tang Y, Pakkenberg B. Marked loss of myelinated nerve fibers in the human brain with age. J Comp Neurol. 2003;462:144–152. doi: 10.1002/cne.10714. [DOI] [PubMed] [Google Scholar]
- Marwarha G, Dasari B, Prasanthi JR, Schommer J, Ghribi O. Leptin reduces the accumulation of Abeta and phosphorylated tau induced by 27-hydroxycholesterol in rabbit organotypic slices. J Alzheimers Dis. 2010;19:1007–1019. doi: 10.3233/JAD-2010-1298. [PubMed: 20157255] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mazzoccoli G, Pazienza V, Piepoli A, Muscarella LA, Inglese M, De Cata A, Giuliani F, Tarquini R. Hypothalamus-hypophysis-thyroid axis function in healthy aging. J Biol Regul Homeost Agents. 2010;24:433–439. [PubMed] [Google Scholar]
- McIntosh AM, Hall J, Lymer GK, Sussmann JE, Lawrie SM. Genetic risk for white matter abnormalities in bipolar disorder. Int Rev Psychiatry. 2009;21:387–393. doi: 10.1080/09540260902962180. [DOI] [PubMed] [Google Scholar]
- McQuillin A, Rizig M, Gurling HM. A microarray gene expression study of the molecular pharmacology of lithium carbonate on mouse brain mRNA to understand the neurobiology of mood stabilization and treatment of bipolar affective disorder. Pharmacogenet Genomics. 2007;17:605–617. doi: 10.1097/FPC.0b013e328011b5b2. [DOI] [PubMed] [Google Scholar]
- Medina M, Wandosell F. Deconstructing GSK-3: The Fine Regulation of Its Activity. Int J Alzheimers Dis. 2011;2011:479249. doi: 10.4061/2011/479249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meltzer HY, Massey BW. The role of serotonin receptors in the action of atypical antipsychotic drugs. Curr Opin Pharmacol. 2011;11:59–67. doi: 10.1016/j.coph.2011.02.007. [DOI] [PubMed] [Google Scholar]
- Miller JS, Tallarida RJ, Unterwald EM. Cocaine-induced hyperactivity and sensitization are dependent on GSK3. Neuropharmacology. 2009;56:1116–1123. doi: 10.1016/j.neuropharm.2009.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller JS, Tallarida RJ, Unterwald EM. Inhibition of GSK3 attenuates dopamine D1 receptor agonist-induced hyperactivity in mice. Brain Res Bull. 2010;82:184–187. doi: 10.1016/j.brainresbull.2010.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitew S, Kirkcaldie MT, Halliday GM, Shepherd CE, Vickers JC, Dickson TC. Focal demyelination in Alzheimer's disease and transgenic mouse models. Acta Neuropathol. 2010;119:567–577. doi: 10.1007/s00401-010-0657-2. [DOI] [PubMed] [Google Scholar]
- Miyamoto Y, Yamauchi J, Chan JR, Okada A, Tomooka Y, Hisanaga S, Tanoue A. Cdk5 regulates differentiation of oligodendrocyte precursor cells through the direct phosphorylation of paxillin. J Cell Sci. 2007;120:4355–4366. doi: 10.1242/jcs.018218. [DOI] [PubMed] [Google Scholar]
- Narayanan SP, Flores AI, Wang F, Macklin WB. Akt signals through the mammalian target of rapamycin pathway to regulate CNS myelination. J Neurosci. 2009;29:6860–6870. doi: 10.1523/JNEUROSCI.0232-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Narimatsu N, Harada N, Kurihara H, Nakagata N, Sobue K, Okajima K. Donepezil improves cognitive function in mice by increasing the production of insulin-like growth factor-I in the hippocampus. J Pharmacol Exp Ther. 2009;330:2–12. doi: 10.1124/jpet.108.147280. [DOI] [PubMed] [Google Scholar]
- Newbern JM, Li X, Shoemaker SE, Zhou J, Zhong J, Wu Y, Bonder D, Hollenback S, Coppola G, Geschwind DH, Landreth GE, Snider WD. Specific functions for ERK/MAPK signaling during PNS development. Neuron. 2011;69:91–105. doi: 10.1016/j.neuron.2010.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicodemus KK, Law AJ, Radulescu E, Luna A, Kolachana B, Vakkalanka R, Rujescu D, Giegling I, Straub RE, McGee K, Gold B, Dean M, Muglia P, Callicott JH, Tan HY, Weinberger DR. Biological validation of increased schizophrenia risk with NRG1, ERBB4, and AKT1 epistasis via functional neuroimaging in healthy controls. Arch Gen Psychiatry. 2011;67:991–1001. doi: 10.1001/archgenpsychiatry.2010.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicolia V, Fuso A, Cavallaro RA, Di Luzio A, Scarpa S. B vitamin deficiency promotes tau phosphorylation through regulation of GSK3beta and PP2A. J Alzheimers Dis. 2011;19:895–907. doi: 10.3233/JAD-2010-1284. [DOI] [PubMed] [Google Scholar]
- Niu J, Mei F, Li N, Wang H, Li X, Kong J, Xiao L. Haloperidol promotes proliferation but inhibits differentiation in rat oligodendrocyte progenitor cell cultures. Biochem Cell Biol. 2010;88:611–620. doi: 10.1139/O09-178. [DOI] [PubMed] [Google Scholar]
- Noble M, Arhin A, Gass D, Mayer-Proschel M. The cortical ancestry of oligodendrocytes: common principles and novel features. Dev Neurosci. 2003;25:217–233. doi: 10.1159/000072270. [DOI] [PubMed] [Google Scholar]
- Nuechterlein KH, Miklowitz DJ, Ventura J, Gitlin MJ, Stoddard M, Lukoff D. Classifying episodes in schizophrenia and bipolar disorder: criteria for relapse and remission applied to recent-onset samples. Psychiatry Res. 2006;144:153–166. doi: 10.1016/j.psychres.2004.04.018. [DOI] [PubMed] [Google Scholar]
- O'Kusky J, Colonnier M. Postnatal changes in the number of astrocytes, oligodendrocytes, and microglia in the visual cortex (area 17) of the macaque monkey: a stereological analysis in normal and monocularly deprived animals. J Comp Neurol. 1982;210:307–315. doi: 10.1002/cne.902100309. [DOI] [PubMed] [Google Scholar]
- Obermayr RP, Mayerhofer L, Knechtelsdorfer M, Mersich N, Huber ER, Geyer G, Tragl KH. The age-related down-regulation of the growth hormone/insulin-like growth factor-1 axis in the elderly male is reversed considerably by donepezil, a drug for Alzheimer's disease. Exp Gerontol. 2005;40:157–163. doi: 10.1016/j.exger.2004.11.001. [DOI] [PubMed] [Google Scholar]
- Pan JQ, Lewis MC, Ketterman JK, Clore EL, Riley M, Richards KR, Berry-Scott E, Liu X, Wagner FF, Holson EB, Neve RL, Biechele TL, Moon RT, Scolnick EM, Petryshen TL, Haggarty SJ. AKT Kinase Activity Is Required for Lithium to Modulate Mood-Related Behaviors in Mice. Neuropsychopharmacology. 2011;36:1397–1411. doi: 10.1038/npp.2011.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parent M, Wallman MJ, Descarries L. Distribution and ultrastructural features of the serotonin innervation in rat and squirrel monkey subthalamic nucleus. Eur J Neurosci. 2010;31:1233–1242. doi: 10.1111/j.1460-9568.2010.07143.x. [DOI] [PubMed] [Google Scholar]
- Pekary AE, Carlson HE, Yamada T, Sharp B, Walfish PG, Hershman JM. Thyrotropinreleasing hormone levels decrease in hypothalamus of aging rats. Neurobiol Aging. 1984;5:221–226. doi: 10.1016/0197-4580(84)90066-6. [DOI] [PubMed] [Google Scholar]
- Pekary AE, Sattin A, Blood J. Rapid modulation of TRH and TRH-like peptide release in rat brain and peripheral tissues by leptin. Brain Res. 2010;1345:9–18. doi: 10.1016/j.brainres.2010.05.039. [DOI] [PubMed] [Google Scholar]
- Pekary AE, Sattin A, RL LL. Electroconvulsive seizures increase levels of pGlu-Glu-Pro-NH2 (EEP) in rat brain. Peptides. 1999;20:107–119. doi: 10.1016/s0196-9781(98)00140-5. [DOI] [PubMed] [Google Scholar]
- Pekary AE, Sattin A, Stevens SA. Rapid modulation of TRH-like peptides in rat brain by thyroid hormones. Peptides. 2006;27:1577–1588. doi: 10.1016/j.peptides.2005.10.015. [DOI] [PubMed] [Google Scholar]
- Peters A. The effects of normal aging on myelinated nerve fibers in monkey central nervous system. Front Neuroanat. 2009;3:11. doi: 10.3389/neuro.05.011.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peters A, Sethares C. Is there remyelination during aging of the primate central nervous system? J Comp Neurol. 2003;460:238–254. doi: 10.1002/cne.10639. [DOI] [PubMed] [Google Scholar]
- Peters A, Sethares C. Oligodendrocytes, their Progenitors and other Neuroglial Cells in the Aging Primate Cerebral Cortex. Cereb Cortex. 2004;14:995–1007. doi: 10.1093/cercor/bhh060. [DOI] [PubMed] [Google Scholar]
- Peters A, Verderosa A, Sethares C. The neuroglial population in the primary visual cortex of the aging rhesus monkey. Glia. 2008;56:1151–1161. doi: 10.1002/glia.20686. [DOI] [PubMed] [Google Scholar]
- Pimlott SL, Piggott M, Owens J, Greally E, Court JA, Jaros E, Perry RH, Perry EK, Wyper D. Nicotinic Acetylcholine Receptor Distribution in Alzheimer's Disease, Dementia with Lewy Bodies, Parkinson's Disease, and Vascular Dementia: In Vitro Binding Study Using 5-[(125)I]-A-85380. Neuropsychopharmacology. 2004;29:108–116. doi: 10.1038/sj.npp.1300302. [DOI] [PubMed] [Google Scholar]
- Pinazo-Duran MD, Pons-Vazquez S, Gallego-Pinazo R, Galbis Estrada C, Zanon-Moreno V, Vila Bou V, Sanz Solana P. Thyroid hormone deficiency disrupts rat eye neurodevelopment. Brain Res. 2011;1392:16–26. doi: 10.1016/j.brainres.2011.04.005. [DOI] [PubMed] [Google Scholar]
- Polter A, Beurel E, Yang S, Garner R, Song L, Miller CA, Sweatt JD, McMahon L, Bartolucci AA, Li X, Jope RS. Deficiency in the inhibitory serine-phosphorylation of glycogen synthase kinase-3 increases sensitivity to mood disturbances. Neuropsychopharmacology. 2010;35:1761–1774. doi: 10.1038/npp.2010.43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Power J, Mayer-Proschel M, Smith J, Noble M. Oligodendrocyte precursor cells from different brain regions express divergent properties consistent with the differing time courses of myelination in these regions. Dev Biol. 2002;245:362–375. doi: 10.1006/dbio.2002.0610. [DOI] [PubMed] [Google Scholar]
- Purves D, Lichtman JW. Elimination of synapses in the developing nervous system. Science. 1980;210:153–157. doi: 10.1126/science.7414326. [DOI] [PubMed] [Google Scholar]
- Raff MC, Lillien LE, Richardson WD, Burne JF, Noble MD. Platelet-derived growth factor from astrocytes drives the clock that times oligodendrocyte development in culture. Nature. 1988;333:562–565. doi: 10.1038/333562a0. [DOI] [PubMed] [Google Scholar]
- Rajkowska G, Halaris A, Selemon LD. Reductions in neuronal and glial density characterize the dorsolateral prefrontal cortex in bipolar disorder. Biol Psychiatry. 2001;49:741–752. doi: 10.1016/s0006-3223(01)01080-0. [DOI] [PubMed] [Google Scholar]
- Regenold WT, Phatak P, Marano CM, Gearhart L, Viens CH, Hisley KC. Myelin staining of deep white matter in the dorsolateral prefrontal cortex in schizophrenia, bipolar disorder, and unipolar major depression. Psychiatry Res. 2007;151:179–188. doi: 10.1016/j.psychres.2006.12.019. [DOI] [PubMed] [Google Scholar]
- Rehani K, Scott DA, Renaud D, Hamza H, Williams LR, Wang H, Martin M. Cotinine-induced convergence of the cholinergic and PI3 kinase-dependent anti-inflammatory pathways in innate immune cells. Biochim Biophys Acta. 2008;1783:375–382. doi: 10.1016/j.bbamcr.2007.12.003. [DOI] [PubMed] [Google Scholar]
- Rinholm JE, Hamilton NB, Kessaris N, Richardson WD, Bergersen LH, Attwell D. Regulation of oligodendrocyte development and myelination by glucose and lactate. J Neurosci. 2011;31:538–548. doi: 10.1523/JNEUROSCI.3516-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson DG, Woerner MG, McMeniman M, Mendelowitz A, Bilder RM. Symptomatic and functional recovery from a first episode of schizophrenia or schizoaffective disorder. Am J Psychiatry. 2004;161:473–479. doi: 10.1176/appi.ajp.161.3.473. [DOI] [PubMed] [Google Scholar]
- Roh MS, Kang UG, Shin SY, Lee YH, Jung HY, Juhnn YS, Kim YS. Biphasic changes in the Ser-9 phosphorylation of glycogen synthase kinase-3beta after electroconvulsive shock in the rat brain. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27:1–5. doi: 10.1016/s0278-5846(02)00307-x. [DOI] [PubMed] [Google Scholar]
- Roosendaal SD, Moraal B, Pouwels PJ, Vrenken H, Castelijns JA, Barkhof F, Geurts JJ. Accumulation of cortical lesions in MS: relation with cognitive impairment. Mult Scler. 2009;15:708–714. doi: 10.1177/1352458509102907. [DOI] [PubMed] [Google Scholar]
- Rutishauser U, Ross IB, Mamelak AN, Schuman EM. Human memory strength is predicted by theta-frequency phase-locking of single neurons. Nature. 2010;464:903–907. doi: 10.1038/nature08860. [DOI] [PubMed] [Google Scholar]
- Salami M, Itami C, Tsumoto T, Kimura F. Change of conduction velocity by regional myelination yields constant latency irrespective of distance between thalamus and cortex. Proc Natl Acad Sci U S A. 2003;100:6174–6179. doi: 10.1073/pnas.0937380100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salat DH, Tuch DS, Greve DN, van der Kouwe AJ, Hevelone ND, Zaleta AK, Rosen BR, Fischl B, Corkin S, Rosas HD, Dale AM. Age-related alterations in white matter microstructure measured by diffusion tensor imaging. Neurobiol Aging. 2005;26:1215–1227. doi: 10.1016/j.neurobiolaging.2004.09.017. [DOI] [PubMed] [Google Scholar]
- Saravanan B, Jacob KS, Johnson S, Prince M, Bhugra D, David AS. Outcome of first-episode schizophrenia in India: longitudinal study of effect of insight and psychopathology. Br J Psychiatry. 2010;196:454–459. doi: 10.1192/bjp.bp.109.068577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sattin A. The role of TRH and related peptides in the mechanism of action of ECT. J Ect. 1999;15:76–92. [PubMed] [Google Scholar]
- Schoonover CM, Seibel MM, Jolson DM, Stack MJ, Rahman RJ, Mariash CN, Anderson GW. Thyroid hormone regulates oligodendrocyte accumulation in developing rat brain white matter tracts. Endocrinology. 2004;145:5013–5020. doi: 10.1210/en.2004-0065. [DOI] [PubMed] [Google Scholar]
- Seeman P. Dopamine D2 receptors as treatment targets in schizophrenia. Clin Schizophr Relat Psychoses. 2010;4:56–73. doi: 10.3371/CSRP.4.1.5. [DOI] [PubMed] [Google Scholar]
- Sei Y, Li Z, Song J, Ren-Patterson R, Tunbridge EM, Iizuka Y, Inoue M, Alfonso BT, Beltaifa S, Nakai Y, Kolachana BS, Chen J, Weinberger DR. Epistatic and functional interactions of catechol-o-methyltransferase (COMT) and AKT1 on neuregulin1-ErbB signaling in cell models. PLoS One. 2010;5:e10789. doi: 10.1371/journal.pone.0010789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Selemon LD, Lidow MS, Goldman-Rakic PS. Increased volume and glial density in primate prefrontal cortex associated with chronic antipsychotic drug exposure. Biol Psychiatry. 1999;46:161–172. doi: 10.1016/s0006-3223(99)00113-4. [DOI] [PubMed] [Google Scholar]
- Selemon LD, Rajkowska G, Goldman-Rakic PS. Abnormally high neuronal density in the schizophrenic cortex. A morphometric analysis of prefrontal area 9 and occipital area 17. Arch Gen Psychiatry. 1995;52:805–818. doi: 10.1001/archpsyc.1995.03950220015005. discussion 819-820. [DOI] [PubMed] [Google Scholar]
- Selemon LD, Rajkowska G, Goldman-Rakic PS. Elevated neuronal density in prefrontal area 46 in brains from schizophrenic patients: application of a three-dimensional, stereologic counting method. J Comp Neurol. 1998;392:402–412. [PubMed] [Google Scholar]
- Shen S, Sandoval J, Swiss VA, Li J, Dupree J, Franklin RJ, Casaccia-Bonnefil P. Agedependent epigenetic control of differentiation inhibitors is critical for remyelination efficiency. Nat Neurosci. 2008;11:1024–1034. doi: 10.1038/nn.2172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siebert JR, Osterhout DJ. Oligodendroglial cells express and secrete reelin. Anat Rec (Hoboken) 2011;294:759–763. doi: 10.1002/ar.21370. [DOI] [PubMed] [Google Scholar]
- Simon B, Schmidt S, Lukas C, Gieseke J, Traber F, Knol DL, Willinek WA, Geurts JJ, Schild HH, Barkhof F, Wattjes MP. Improved in vivo detection of cortical lesions in multiple sclerosis using double inversion recovery MR imaging at 3 Tesla. Eur Radiol. 2010;20:1675–1683. doi: 10.1007/s00330-009-1705-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simon C, Gotz M, Dimou L. Progenitors in the adult cerebral cortex: Cell cycle properties and regulation by physiological stimuli and injury. Glia. 2011;59:869–881. doi: 10.1002/glia.21156. [DOI] [PubMed] [Google Scholar]
- Smaers JB, Steele J, Case CR, Cowper A, Amunts K, Zilles K. Primate prefrontal cortex evolution: human brains are the extreme of a lateralized ape trend. Brain Behav Evol. 2011;77:67–78. doi: 10.1159/000323671. [DOI] [PubMed] [Google Scholar]
- Smiley JF, Levey AI, Ciliax BJ, Goldman-Rakic PS. D1 dopamine receptor immunoreactivity in human and monkey cerebral cortex: predominant and extrasynaptic localization in dendritic spines. Proc Natl Acad Sci U S A. 1994;91:5720–5724. doi: 10.1073/pnas.91.12.5720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith RS, Koles ZJ. Myelinated nerve fibers: computed effect of myelin thickness on conduction velocity. Am J Physiol. 1970;219:1256–1258. doi: 10.1152/ajplegacy.1970.219.5.1256. [DOI] [PubMed] [Google Scholar]
- Sole B, Martinez-Aran A, Torrent C, Bonnin CM, Reinares M, Popovic D, Sanchez-Moreno J, Vieta E. Are bipolar II patients cognitively impaired? A systematic review. Psychol Med. 2011:1–13. doi: 10.1017/S0033291711000018. [DOI] [PubMed] [Google Scholar]
- Solowij N, Yucel M, Respondek C, Whittle S, Lindsay E, Pantelis C, Lubman DI. Cerebellar white-matter changes in cannabis users with and without schizophrenia. Psychol Med. 2011;41:2349–2359. doi: 10.1017/S003329171100050X. [DOI] [PubMed] [Google Scholar]
- Souza BR, Romano-Silva MA, Tropepe V. Dopamine D2 receptor activity modulates Akt signaling and alters GABAergic neuron development and motor behavior in zebrafish larvae. J Neurosci. 2011;31:5512–5525. doi: 10.1523/JNEUROSCI.5548-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spilman P, Podlutskaya N, Hart MJ, Debnath J, Gorostiza O, Bredesen D, Richardson A, Strong R, Galvan V. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer's disease. PLoS One. 2010;5:e9979. doi: 10.1371/journal.pone.0009979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spittaels K, Van den Haute C, Van Dorpe J, Terwel D, Vandezande K, Lasrado R, Bruynseels K, Irizarry M, Verhoye M, Van Lint J, Vandenheede JR, Ashton D, Mercken M, Loos R, Hyman B, Van der Linden A, Geerts H, Van Leuven F. Neonatal neuronal overexpression of glycogen synthase kinase-3 beta reduces brain size in transgenic mice. Neuroscience. 2002;113:797–808. doi: 10.1016/s0306-4522(02)00236-1. [DOI] [PubMed] [Google Scholar]
- Stark AK, Uylings HB, Sanz-Arigita E, Pakkenberg B. Glial cell loss in the anterior cingulate cortex, a subregion of the prefrontal cortex, in subjects with schizophrenia. Am J Psychiatry. 2004;161:882–888. doi: 10.1176/appi.ajp.161.5.882. [DOI] [PubMed] [Google Scholar]
- Stranahan AM, Salas-Vega S, Jiam NT, Gallagher M. Interference with reelin signaling in the lateral entorhinal cortex impairs spatial memory. Neurobiol Learn Mem. 2011;96:150–155. doi: 10.1016/j.nlm.2011.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Subotnik KL, Nuechterlein KH, Ventura J, Gitlin MJ, Marder SR, Mintz J, Hellemann GS, Thornton LA, Singh IR. Risperidone nonadherence and return of positive symptoms in the early course of schizophrenia. Am J Psychiatry. 2011;168:286–292. doi: 10.1176/appi.ajp.2010.09010087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sutherland C. What Are the bona fide GSK3 Substrates? Int J Alzheimers Dis. 2011;2011:505607. doi: 10.4061/2011/505607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szuba MP, Amsterdam JD, Fernando AT, 3rd, Gary KA, Whybrow PC, Winokur A. Rapid antidepressant response after nocturnal TRH administration in patients with bipolar type I and bipolar type II major depression. J Clin Psychopharmacol. 2005;25:325–330. doi: 10.1097/01.jcp.0000169037.17884.79. [DOI] [PubMed] [Google Scholar]
- Tamura M, Nakamura M, Ogawa Y, Toyama Y, Miura M, Okano H. Targeted expression of antiapoptotic protein p35 in oligodendrocytes reduces delayed demyelination and functional impairment after spinal cord injury. Glia. 2005;51:312–321. doi: 10.1002/glia.20212. [DOI] [PubMed] [Google Scholar]
- Tei E, Yamamoto H, Watanabe T, Miyazaki A, Nakadate T, Kato N, Mimura M. Use of serum insulin-like growth factor-I levels to predict psychiatric non-response to donepezil in patients with Alzheimer's disease. Growth Horm IGF Res. 2008;18:47–54. doi: 10.1016/j.ghir.2007.07.006. [DOI] [PubMed] [Google Scholar]
- Teixeira CM, Martin ED, Sahun I, Masachs N, Pujadas L, Corvelo A, Bosch C, Rossi D, Martinez A, Maldonado R, Dierssen M, Soriano E. Overexpression of Reelin Prevents the Manifestation of Behavioral Phenotypes Related to Schizophrenia and Bipolar Disorder. Neuropsychopharmacology. 2011;36:2395–2405. doi: 10.1038/npp.2011.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thiselton DL, Vladimirov VI, Kuo PH, McClay J, Wormley B, Fanous A, O'Neill FA, Walsh D, Van den Oord EJ, Kendler KS, Riley BP. AKT1 is associated with schizophrenia across multiple symptom dimensions in the Irish study of high density schizophrenia families. Biol Psychiatry. 2008;63:449–457. doi: 10.1016/j.biopsych.2007.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomas GM, Frame S, Goedert M, Nathke I, Polakis P, Cohen P. A GSK3-binding peptide from FRAT1 selectively inhibits the GSK3-catalysed phosphorylation of axin and beta-catenin. FEBS Lett. 1999;458:247–251. doi: 10.1016/s0014-5793(99)01161-8. [DOI] [PubMed] [Google Scholar]
- Thornton TM, Pedraza-Alva G, Deng B, Wood CD, Aronshtam A, Clements JL, Sabio G, Davis RJ, Matthews DE, Doble B, Rincon M. Phosphorylation by p38 MAPK as an alternative pathway for GSK3beta inactivation. Science. 2008;320:667–670. doi: 10.1126/science.1156037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tiihonen J, Haukka J, Taylor M, Haddad PM, Patel MX, Korhonen P. A nationwide cohort study of oral and depot antipsychotics after first hospitalization for schizophrenia. Am J Psychiatry. 2011;168:603–609. doi: 10.1176/appi.ajp.2011.10081224. [DOI] [PubMed] [Google Scholar]
- Tobias MC, O'Neill J, Hudkins M, Bartzokis G, Dean AC, London ED. White-matter abnormalities in brain during early abstinence from methamphetamine abuse. Psychopharmacology (Berl) 2010;209:13–24. doi: 10.1007/s00213-009-1761-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tong XP, Li XY, Zhou B, Shen W, Zhang ZJ, Xu TL, Duan S. Ca(2+) signaling evoked by activation of Na(+) channels and Na(+)/Ca(2+) exchangers is required for GABA-induced NG2 cell migration. J Cell Biol. 2009;186:113–128. doi: 10.1083/jcb.200811071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torrey EF, Barci BM, Webster MJ, Bartko JJ, Meador-Woodruff JH, Knable MB. Neurochemical markers for schizophrenia, bipolar disorder, and major depression in postmortem brains. Biol Psychiatry. 2005;57:252–260. doi: 10.1016/j.biopsych.2004.10.019. [DOI] [PubMed] [Google Scholar]
- Trivedi MH, Greer TL, Church TS, Carmody TJ, Grannemann BD, Galper DI, Dunn AL, Earnest CP, Sunderajan P, Henley SS, Blair SN. Exercise as an augmentation treatment for nonremitted major depressive disorder: a randomized, parallel dose comparison. J Clin Psychiatry. 2011;72:677–684. doi: 10.4088/JCP.10m06743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tyler WA, Gangoli N, Gokina P, Kim HA, Covey M, Levison SW, Wood TL. Activation of the mammalian target of rapamycin (mTOR) is essential for oligodendrocyte differentiation. J Neurosci. 2009;29:6367–6378. doi: 10.1523/JNEUROSCI.0234-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uhlhaas PJ, Singer W. Abnormal neural oscillations and synchrony in schizophrenia. Nat Rev Neurosci. 2010;11:100–113. doi: 10.1038/nrn2774. [DOI] [PubMed] [Google Scholar]
- Umbriaco D, Garcia S, Beaulieu C, Descarries L. Relational features of acetylcholine, noradrenaline, serotonin and GABA axon terminals in the stratum radiatum of adult rat hippocampus (CA1) Hippocampus. 1995;5:605–620. doi: 10.1002/hipo.450050611. [DOI] [PubMed] [Google Scholar]
- Uranova NA, Vikhreva OV, Rachmanova VI, Orlovskaya DD. Ultrastructural alterations of myelinated fibers and oligodendrocytes in the prefrontal cortex in schizophrenia: A postmortem morphometric study. Schizophrenia Research and Treatment. 2011;2011 doi: 10.1155/2011/325789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uranova NA, Vostrikov VM, Orlovskaya DD, Rachmanova VI. Oligodendroglial density in the prefrontal cortex in schizophrenia and mood disorders: a study from the Stanley Neuropathology Consortium. Schizophr Res. 2004;67:269–275. doi: 10.1016/S0920-9964(03)00181-6. [DOI] [PubMed] [Google Scholar]
- van der Schot AC, Vonk R, Brans RG, van Haren NE, Koolschijn PC, Nuboer V, Schnack HG, van Baal GC, Boomsma DI, Nolen WA, Hulshoff Pol HE, Kahn RS. Influence of genes and environment on brain volumes in twin pairs concordant and discordant for bipolar disorder. Arch Gen Psychiatry. 2009;66:142–151. doi: 10.1001/archgenpsychiatry.2008.541. [DOI] [PubMed] [Google Scholar]
- Van't Veer A, Du Y, Fischer TZ, Boetig DR, Wood MR, Dreyfus CF. Brain-derived neurotrophic factor effects on oligodendrocyte progenitors of the basal forebrain are mediated through trkB and the MAP kinase pathway. J Neurosci Res. 2009;87:69–78. doi: 10.1002/jnr.21841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vaupel DB, Tella SR, Huso DL, Wagner VO, 3rd, Mukhin AG, Chefer SI, Horti AG, London ED, Koren AO, Kimes AS. Pharmacological and Toxicological Evaluation of 2-Fluoro-3-(2(S)-azetidinylmethoxy)pyridine (2-F-A-85380), a Ligand for Imaging Cerebral Nicotinic Acetylcholine Receptors with Positron Emission Tomography. J Pharmacol Exp Ther. 2005;312:355–365. doi: 10.1124/jpet.104.073999. [DOI] [PubMed] [Google Scholar]
- Velez-Fort M, Maldonado PP, Butt AM, Audinat E, Angulo MC. Postnatal switch from synaptic to extrasynaptic transmission between interneurons and NG2 cells. J Neurosci. 2010;30:6921–6929. doi: 10.1523/JNEUROSCI.0238-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Venkatasubramanian G, Chittiprol S, Neelakantachar N, Shetty T, Gangadhar BN. Effect of antipsychotic treatment on Insulin-like Growth Factor-1 and cortisol in schizophrenia: a longitudinal study. Schizophr Res. 2010;119:131–137. doi: 10.1016/j.schres.2010.01.033. [DOI] [PubMed] [Google Scholar]
- Venneri A, Lane R. Effects of cholinesterase inhibition on brain white matter volume in Alzheimer's disease. Neuroreport. 2009;20:285–288. doi: 10.1097/wnr.0b013e3283207d21. [DOI] [PubMed] [Google Scholar]
- Vostrikov V, Uranova N. Age-related increase in the number of oligodendrocytes is dysregulated in schizophrenia and mood disorders. Schizophrenia Research and Treatment. 2011;2011 doi: 10.1155/2011/174689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vostrikov VM, Uranova NA, Orlovskaya DD. Deficit of perineuronal oligodendrocytes in the prefrontal cortex in schizophrenia and mood disorders. Schizophr Res. 2007;94:273–280. doi: 10.1016/j.schres.2007.04.014. [DOI] [PubMed] [Google Scholar]
- Wallis M. Molecular evolution of the thyrotrophin-releasing hormone precursor in vertebrates: insights from comparative genomics. J Neuroendocrinol. 2010;22:608–619. doi: 10.1111/j.1365-2826.2010.01996.x. [DOI] [PubMed] [Google Scholar]
- Wang H, Brown J, Gu Z, Garcia CA, Liang R, Alard P, Beurel E, Jope RS, Greenway T, Martin M. Convergence of the mammalian target of rapamycin complex 1- and glycogen synthase kinase 3-beta-signaling pathways regulates the innate inflammatory response. J Immunol. 2011a;186:5217–5226. doi: 10.4049/jimmunol.1002513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Brown J, Martin M. Glycogen synthase kinase 3: a point of convergence for the host inflammatory response. Cytokine. 2011b;53:130–140. doi: 10.1016/j.cyto.2010.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Xu H, Niu J, Mei F, Li X, Kong J, Cai W, Xiao L. Haloperidol activates quiescent oligodendroglia precursor cells in the adult mouse brain. Schizophr Res. 2010;119:164–174. doi: 10.1016/j.schres.2010.02.1068. [DOI] [PubMed] [Google Scholar]
- Wedenoja J, Tuulio-Henriksson A, Suvisaari J, Loukola A, Paunio T, Partonen T, Varilo T, Lonnqvist J, Peltonen L. Replication of Association Between Working Memory and Reelin, a Potential Modifier Gene in Schizophrenia. Biol Psychiatry. 2010;67:983–991. doi: 10.1016/j.biopsych.2009.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen Y, Planel E, Herman M, Figueroa HY, Wang L, Liu L, Lau LF, Yu WH, Duff KE. Interplay between cyclin-dependent kinase 5 and glycogen synthase kinase 3beta mediated by neuregulin signaling leads to differential effects on tau phosphorylation and amyloid precursor protein processing. J Neurosci. 2008;28:2624–2632. doi: 10.1523/JNEUROSCI.5245-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wennstrom M, Hellsten J, Ekdahl CT, Tingstrom A. Electroconvulsive seizures induce proliferation of NG2-expressing glial cells in adult rat hippocampus. Biol Psychiatry. 2003;54:1015–1024. doi: 10.1016/s0006-3223(03)00693-0. [DOI] [PubMed] [Google Scholar]
- Wennstrom M, Hellsten J, Tingstrom A. Electroconvulsive seizures induce proliferation of NG2-expressing glial cells in adult rat amygdala. Biol Psychiatry. 2004;55:464–471. doi: 10.1016/j.biopsych.2003.11.011. [DOI] [PubMed] [Google Scholar]
- White T, Magnotta VA, Bockholt HJ, Williams S, Wallace S, Ehrlich S, Mueller BA, Ho BC, Jung RE, Clark VP, Lauriello J, Bustillo JR, Schulz SC, Gollub RL, Andreasen NC, Calhoun VD, Lim KO. Global white matter abnormalities in schizophrenia: a multisite diffusion tensor imaging study. Schizophr Bull. 2011;37:222–232. doi: 10.1093/schbul/sbp088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams S, Boksa P. Gamma oscillations and schizophrenia. J Psychiatry Neurosci. 2010;35:75–77. doi: 10.1503/jpn.100021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xi D, Li YC, Snyder MA, Gao RY, Adelman AE, Zhang W, Shumsky JS, Gao WJ. Group II Metabotropic Glutamate Receptor Agonist Ameliorates MK801-Induced Dysfunction of NMDA Receptors via the Akt/GSK-3beta Pathway in Adult Rat Prefrontal Cortex. Neuropsychopharmacology. 2011;36:1260–1274. doi: 10.1038/npp.2011.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu H, Yang HJ, McConomy B, Browning R, Li XM. Behavioral and neurobiological changes in C57BL/6 mouse exposed to cuprizone: effects of antipsychotics. Front Behav Neurosci. 2010;4:8. doi: 10.3389/fnbeh.2010.00008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang H-J, Wang L, Cheng Q, Xu H. Abnormal behaviors and micro-structural changes in white matter of juvenile mice repeatedly exposed to amphetamine. Schizophrenia Research and Treatment. 2011;2011 doi: 10.1155/2011/542896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye P, Hu Q, Liu H, Yan Y, D'Ercole AJ. beta-catenin mediates insulin-like growth factor-I actions to promote cyclin D1 mRNA expression, cell proliferation and survival in oligodendroglial cultures. Glia. 2010;58:1031–1041. doi: 10.1002/glia.20984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu M, Narayanan SP, Wang F, Morse E, Macklin WB, Peachey NS. Visual abnormalities associated with enhanced optic nerve myelination. Brain Res. 2011;1374:36–42. doi: 10.1016/j.brainres.2010.12.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu Y, Kastin AJ, Pan W. Reciprocal interactions of insulin and insulin-like growth factor I in receptor-mediated transport across the blood-brain barrier. Endocrinology. 2006;147:2611–2615. doi: 10.1210/en.2006-0020. [DOI] [PubMed] [Google Scholar]
- Yuan X, Eisen AM, McBain CJ, Gallo V. A role for glutamate and its receptors in the regulation of oligodendrocyte development in cerebellar tissue slices. Development. 1998;125:2901–2914. doi: 10.1242/dev.125.15.2901. [DOI] [PubMed] [Google Scholar]
- Zanelli J, Reichenberg A, Morgan K, Fearon P, Kravariti E, Dazzan P, Morgan C, Zanelli C, Demjaha A, Jones PB, Doody GA, Kapur S, Murray RM. Specific and generalized neuropsychological deficits: a comparison of patients with various first-episode psychosis presentations. Am J Psychiatry. 2010;167:78–85. doi: 10.1176/appi.ajp.2009.09010118. [DOI] [PubMed] [Google Scholar]
- Zhao WQ, Townsend M. Insulin resistance and amyloidogenesis as common molecular foundation for type 2 diabetes and Alzheimer's disease. Biochim Biophys Acta. 2009;1792:482–496. doi: 10.1016/j.bbadis.2008.10.014. [DOI] [PubMed] [Google Scholar]
- Zhao Z, Ksiezak-Reding H, Riggio S, Haroutunian V, Pasinetti GM. Insulin receptor deficits in schizophrenia and in cellular and animal models of insulin receptor dysfunction. Schizophr Res. 2006;84:1–14. doi: 10.1016/j.schres.2006.02.009. [DOI] [PubMed] [Google Scholar]
- Zhuo JM, Portugal GS, Kruger WD, Wang H, Gould TJ, Pratico D. Diet-induced hyperhomocysteinemia increases amyloid-beta formation and deposition in a mouse model of Alzheimer's disease. Curr Alzheimer Res. 2011;7:140–149. doi: 10.2174/156720510790691326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ziskin JL, Nishiyama A, Rubio M, Fukaya M, Bergles DE. Vesicular release of glutamate from unmyelinated axons in white matter. Nat Neurosci. 2007;10:321–330. doi: 10.1038/nn1854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 2011;12:21–35. doi: 10.1038/nrm3025. [DOI] [PMC free article] [PubMed] [Google Scholar]



