Sei sulla pagina 1di 28

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/285088812

Neurodevelopment, neuroplasticity, and new genes for schizophrenia.


Progress in

Article  in  Brain Research · January 2005

CITATIONS READS
42 84

3 authors, including:

Steven Arnold Konrad Talbot


Harvard Medical School University of California, Los Angeles
359 PUBLICATIONS   17,039 CITATIONS    59 PUBLICATIONS   4,076 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Brain Insulin Resistance and Its Treatment in Neurodegenerative Disorders View project

Neurobiology of schizophrenia and antipsychotic effects View project

All content following this page was uploaded by Konrad Talbot on 09 February 2017.

The user has requested enhancement of the downloaded file.


Progress in Brain Research, Vol. 147
ISSN 0079-6123
Copyright ß 2005 Elsevier BV. All rights reserved

CHAPTER 23

Neurodevelopment, neuroplasticity, and new genes


for schizophrenia

Steven E. Arnold*, Konrad Talbot and Chang-Gyu Hahn

Cellular and Molecular Neuropathology Program, Center for Neurobiology and Behavior, Department of Psychiatry,
University of Pennsylvania, Philadelphia, PA 19104, USA

Abstract: Schizophrenia is a complex, debilitating neuropsychiatric disorder. Epidemiological, clinical, neuropsycho-


logical, and neurophysiological studies have provided substantial evidence that abnormalities in brain development and
ongoing neuroplasticity play important roles in the pathogenesis of the disorder. Complementing these clinical studies,
a range of cytoarchitectural, morphometric, ultrastructural, immunochemical, and gene expression methods have been
applied in investigations of postmortem brain tissues to characterize the cellular and molecular profile of putative
developmental and plastic abnormalities in schizophrenia. While findings have been diverse and many are in need of
replication, investigations focusing on higher cortical and limbic brain regions are increasingly demonstrating
abnormalities in the structural and molecular integrity of the synaptic complex as well as glutamate-related receptors
and signal transduction pathways that play critical roles in brain development, synaptogenesis, and synaptic plasticity.
Most exciting have been recent associations of schizophrenia with specific genes, such as neuregulin-1, dysbindin-1, and
AKT-1, which are vital to synaptic development, neurotransmission, and plasticity.

Introduction schizophrenia commit suicide (Hafner and Heiden,


2003).
Schizophrenia is a severe, chronic mental illness Schizophrenia is best recognized by the presence of
with a lifetime prevalence of 0.12–1.6% worldwide the so-called ‘‘positive’’ symptoms of schizophrenia
(Goldner et al., 2002). Diagnosis is typically (Eaton et al., 1995; Hafner and Heiden, 2003), which
established in late adolescence or early adulthood, include hallucinations, delusions, and thought dis-
when the disease’s characteristic psychiatric features order. Less conspicuous, though more disabling are
become evident and it often leads to a lifetime of the ‘‘negative’’ and cognitive symptoms of schizo-
suffering for both patient and family. In a recent phrenia (Eaton et al., 1995; Hafner and Heiden, 2003),
World Health Organization report on the burden of which include apathy, social withdrawal, flatness of
disease in developed countries, schizophrenia ranked emotion and affect, and anhedonia that constitute the
fifth among all illnesses in the number of years lost to real deterioration in personality that occurs with the
disability (Murray and Lopez, 1996). It is associated disease. Further, as discussed in greater detail below,
with significant mortality as 4–14% of people with there has been increasing recognition of significant
cognitive deficits in persons with schizophrenia,
especially in attention, memory, and executive
*Corresponding author. Tel.: +1-215-573-2840; functions (Gur et al., 2000; Goldberg et al., 2003)
Fax: +1-215-573-6382; E-mail: sarnold@med.upenn.edu that also contribute greatly to disability. Treatment

DOI: 10.1016/S0079-6123(04)47023-X 319


320

with dopamine-antagonist medications can be very neurodevelopment and neuroplasticity in the patho-
effective in eliminating or diminishing the positive genesis of schizophrenia, and highlight the potential
symptoms of schizophrenia, but to date, medications roles in these processes of several genes that have
have shown little ‘‘if any’’ benefit for the negative and been recently associated with the illness.
cognitive symptoms (Miyamoto et al., 2003).
The pathogenesis of schizophrenia as a brain
disorder has been a matter of intense scrutiny for Clinical and epidemiological evidence supporting
over a century. Emil Kraepelin’s initial formulation aberrant neurodevelopment in schizophrenia
of schizophrenia as ‘‘dementia praecox’’ highlighted
deteriorative aspects of the disease and raised the Epidemiological studies have identified a variety of
possibility of neurodegeneration as the cause prenatal and perinatal factors conferring increased
(Kraepelin, 1919). Supporting that notion is the risk for the later development of schizophrenia via
clear behavioral, cognitive and functional deteriora- deleterious effects on brain development. Many
tion with disease onset, which in some patients studies have found that winter and spring births are
continue and culminate in a state resembling 5–8% more frequent among persons with schizo-
neurodegenerative dementias in late life. However, phrenia than the general population (Torrey et al.,
almost without exception, postmortem studies have 1997). How season of birth affects the brain and risk
failed to find any increase in gliosis, apoptosis, for mental illness is not clear, although the leading
neurodegenerative disease lesions (e.g., amyloid speculation is that infections, more common in winter
plaques, neurofibrillary tangles, Lewy bodies, or and spring, may affect brain development. Mednick
other filamentous inclusions), or other indicators of (1988) found an increased frequency of schizophrenia
neuronal death or injury (e.g., ubiquitination), even in children of mothers who were in their second
in elderly, severely regressed persons with schizo- trimester of pregnancy during the 1957 influenza
phrenia (Arnold et al., 1998; Harrison, 1999). epidemic, although subsequent studies questioned
On the other hand, presence of neurodevelop- such an association (Brown and Susser, 2002).
mental anomalies in schizophrenia has long been Elevated maternal serum immunoglobulin levels
recognized. Hecker, Kraepelin, and Bleuler all near birth (Buka et al., 2001), as well as increased
commented on premorbid physical, mental, and frequency of maternal rubella infection among
behavioral abnormalities (Hecker, 1871; Kraepelin, individuals who later develop schizophrenia (Brown
1919; Bleuler, 1924; Marenco and Weinberger, 2000). et al., 2001), have also been reported and suggest
Contemporary clinical and neurobiological evidence a relationship with intrauterine infection.
increasingly support the role of abnormal neuro- Urban birth may be another epidemiological risk
development in the pathogenesis of schizophrenia factor for schizophrenia. Several studies have found
(Arnold and Rioux, 2001; Weinberger and Marenco, an increase in schizophrenia among those born in
2003). Data from neuropsychological, functional urban environments (Marcelis et al., 1998; Mortensen
neuroimaging, electrophysiological, genetic, and et al., 1999). The mechanism for this is not clear, but
molecular neuropathological studies also suggest possibilities include infectious disease, toxic expo-
abnormalities in neuroplasticity, impairing the sures, malnutrition, elevated density of families with
adaptive capacity of multiple neural systems to a history of mental illness in cities, or ill-defined
respond to environmental stimuli and experience in deleterious effects of growing up in an urban
schizophrenia. The effects of normal maturation and environment. Severe maternal malnutrition has been
aging processes interacting with schizophrenia’s invoked in a study of children conceived during the
developmentally abnormal circuitry and reduced severe Dutch Hunger Winter of 1944–1945. Such
neuroplasticity may lead to the cognitive and children had a two-fold increase in risk for
behavioral deterioration seen during schizophrenia schizophrenia (Susser et al., 1996).
onset and then continuing over the lifespan. In this Perinatal and obstetrical complications may affect
chapter, we review and consider current clinical the brain and have been associated with a higher risk
and neuropathological data that implicate abnormal of schizophrenia. In a meta-analysis by Geddes and
321

Lawries (1995), the risk was two-fold. Among the forms of mental retardation and other brain
complications cited are hypoxemia (e.g., associated disorders (Waldrop et al., 1968; Lohr and Flynn,
with abnormal birth presentation, complicated 1993; Sivkov and Akabaliev, 2003).
Caesarean section), perinatal viral exposure, pre- A neurodevelopmental etiology of schizophrenia
maturity, and ABO blood type incompatibility. is also supported by investigations of intellectual,
A number of minor physical anomalies that are motor, and social development during childhood and
more common in schizophrenia may be found in adolescence. Impaired language acquisition, lower IQ
association with developmental brain abnormalities scores, lower cognitive functioning, poor academic
(O’Callaghan et al., 1991; Schiffman et al., 2002; achievement, and attention deficit disorder have been
Gourion et al., 2004). The most obvious anomaly found in both younger and older school-age children
is head circumference which is a direct correlate of who later develop schizophrenia in adolescence or
brain growth in neonates and children. Several adulthood (Chua and Murray, 1996; Davidson et al.,
studies have reported decreased head circumference 1999; Niemi et al., 2003). Motor abnormalities during
at the time of birth in individuals who later develop childhood associated with schizophrenia include
schizophrenia suggesting impaired or delayed brain delayed development of sitting, standing, and
growth, although methodological issues in these walking, as well as increased occurrence of unusual
studies remain (McNeil et al., 1996; Cantor-Graae movements such as choreoathetotic posturing (Fish
et al., 1998; Cannon et al., 2002). Small adult head et al., 1992; Jones et al., 1994; Walker, 1994; Niemi
circumference has been reported too, although this is et al., 2003).
controversial and at best, subtle. Lastly, abnormalities in social development during
While not directly measuring the trajectory of childhood have been reported by a number of
brain growth, ultimate brain volume is best measured investigators (Niemi et al., 2003). Among the social
with magnetic resonance imaging, and there have adjustment anomalies described are a tendency to
been numerous studies using this technology. Many play alone, poor social confidence, social anxiety,
report increased volume of the lateral ventricles and/ social withdrawal, inappropriate and disruptive
or decreased volume of the superior temporal gyrus, social behaviors, fewer social relations, hypersensi-
hippocampal formation, and parahippocampal gyrus tivity, and a paucity of dating experience (Done et al.,
(McCarley et al., 1999; Talbot and Arnold, 2002; 1994; Jones et al., 1994; Malmberg et al., 1998; Olin
Liddle and Pantelis, 2003). Reductions in gray matter et al., 1998; Davidson et al., 1999; Niemi et al., 2003).
volume and in the volumes of the hippocampal While these behaviors may reflect neurodevelopmen-
formation and parahippocampal gyrus have been tal abnormalities that are inherent in the schizo-
reported at first presentation before neuroleptic phrenia disease process, they may also represent
treatment (Ohnuma et al., 1997; Gur et al., 1999; environmental psychological stressors that foster the
Joyal et al., 2002). Volume changes are less development of mental illness. Our understanding of
consistently found in other brain areas, including the neural basis of social-affiliative behaviors is in its
the prefrontal cortex, cingulate gyrus, parietal lobe, infancy, but it is interesting to note that social-
thalamus, basal ganglia, cerebellar vermis, and affiliative behaviors are entering the field of research
olfactory bulbs (McCarley et al., 1999; Turetsky as target behaviors in mouse models of schizophrenia
et al., 2000b; Liddle and Pantelis, 2003). (Ellenbroek and Cools, 2000; Miyakawa et al., 2003).
The ectodermal germ layer gives rise to the brain
and spinal cord, as well as skin and its many external
physical features. The increased frequency of minor Clinical evidence supporting aberrant neural
physical anomalies in schizophrenia (O’Callaghan plasticity in schizophrenia
et al., 1991; Schiffman et al., 2002; Gourion et al.,
2004), such as a high-arched palate, low-set ears, ‘‘Neuroplasticity’’ is a broad term for any adaptive
anomalous skin ridge patterns, and curved fingers are changes in structure or function of the nervous
thought to derive from abnormal ectodermal devel- system during development, cognition, recovery from
opment and are seen in association with various injury, and aging. In our discussion of neural
322

plasticity in schizophrenia, we will use the term more both positron emission tomography (Meyer-
narrowly to refer to the molecular, chemical, Lindenberg et al., 2001; Abi-Dargham et al., 2002;
morphological, and physiological modifiability of Buchsbaum et al., 2002; Potkin et al., 2002; Abi-
neurons and neurotransmission that occurs with Dargham and Moore, 2003; Erritzoe et al., 2003;
sensory and cognitive neural activity. Kim et al., 2003; Paradiso et al., 2003; Hazlett et al.,
Sensation, perception, attention, memory, learn- 2004) and functional MRI (Menon et al., 2001; Wible
ing, and thought represent the diverse domains of et al., 2001; Gur et al., 2002; van Veen and Carter,
information processing in which deficits have been 2002; Hofer et al., 2003; Jessen et al., 2003;
reported in schizophrenia (Goldberg et al., 2003). The MacDonald and Carter, 2003; Weiss et al., 2003),
deficits span sensory modalities and may reflect a investigations have documented performance deficits
fundamental disturbance in neurobiological mecha- correlated with abnormal brain activation in the
nisms by which the brain responds to and changes ventromedial temporal lobe, prefrontal cortices,
with environmental input. Within the context of limbic subcortical nuclei, and other brain regions
global cognitive impairments in schizophrenia, known to mediate memory, executive functioning,
specific deficits are found in attention, working attention, and emotional processing. These systems
memory, executive function, and episodic memory are characterized by dynamic plasticity, a high degree
(Saykin et al., 1991; Goldberg et al., 2003). Several of interconnectivity and vulnerability to insult. If
brain systems are implicated by these deficits. The fundamental mechanisms of structural, molecular, or
attention processing circuitry is complex and includes physiological plasticity are abnormal in schizophre-
the prefrontal cortex and structures directly con- nia, then these may be evident throughout the CNS,
nected with it, especially the posterior parietal cortex, but most prominent in the highly plastic neural
anterior cingulate gyrus, hippocampal formation, systems that mediate activity-dependent cognitive
and striatum (Fernandez-Duque and Posner, 2001; processes such as attention, memory, and executive
Wall and Messier, 2001; Faw, 2003; Pessoa et al., functioning.
2003; Saint-Cyr, 2003). Deficits in working and Electrophysiological studies have shown abnormal
episodic memory also implicate the prefrontal cortex information processing in schizophrenia by analyzing
(Buckner et al., 2000; Faw, 2003) and its connec- event-related potentials (ERPs) triggered by sensory
tions with the posterior parietal cortex, anterior stimuli. Such ERP abnormalities are common in the
cingulate, temporal and parahippocampal cortices, disorder and include deficits in early preattentive as
and striatum (Andres, 2003). well as late stages of information processing (Erwin
A particularly interesting sensory modality in et al., 1998; Karoumi et al., 2000; Kathmann et al.,
which deficits have been described in schizophrenia 2000; Fallgatter, 2001; Pekkonen et al., 2002;
is olfaction. Many studies over the last decade have McNeely et al., 2003; Turetsky et al., 2003;
reported significant deficits in odor identification, Clementz et al., 2004). Aberrations in encoding of
detection, memory, and hedonics (Moberg et al., input may reflect difficulty in screening out informa-
1999; Moberg and Turetsky, 2003). From the tion and inhibiting responses to irrelevant stimuli.
perspective of neuroplasticity, olfactory system There may be corresponding deficits in response
deficits may be especially instructive because they facilitation to salient stimuli. Three ERP phenomena
could reflect abnormal dynamics in the ongoing have been the focus of investigation on successive
neurogenesis and synaptic turnover that distinguishes stages of stimulus processing and their neural
the primary olfactory structures (olfactory epithelium mechanisms. Failure of stimulus-induced inhibitory
and bulb) from the rest of the brain (Schwob, 2002). processes is evident from studies of the P50 response
Initial studies have described abnormalities in the to paired clicks (Freedman et al., 1991; Erwin et al.,
lineage of olfactory epithelium neurons in schizo- 1998) and on inhibition of an acoustic startle
phrenia (Arnold et al., 2001). response by a preceding sound (i.e., prepulse
Functional neuroimaging during activation by inhibition) (Braff et al., 1992, 2001). In the paired
cognitive or other neurobehavioral tasks has helped click paradigm, the amplitude of the P50 ERP to the
define the pathophysiology of schizophrenia. Using second of two auditory clicks is reduced in healthy
323

people, but not in individuals with schizophrenia. cytoarchitecture, neuronal morphometry, and inner-
Similarly, in the prepulse inhibition paradigm, the vation patterns in an ‘‘archaeological’’ manner to
startle response to a loud stimulus, preceded by a infer the timing and nature of an abnormality in
low-intensity click, is reduced in controls, but not neuronal migration, wiring, or other developmental
patients. These two lines of research indicate a process from the anatomical appearance of the brain
fundamental neural failure to modify electrophysio- at the time of death. In this manner, investigators
logical response to a second stimulus paired to an have reported a number of abnormalities in schizo-
earlier one. Patients with schizophrenia also show phrenia, although most remain controversial.
lower amplitudes of later ERPs, including the N200 Kovelman and Scheibel (1984) and later Conrad
during mismatch negativity conditions using deviant et al. (1991) reported that pyramidal neurons within
tones on a passive background of tones (Hirayasu the subfields of the hippocampus showed greater
et al., 1998; Williams et al., 2000; Brown et al., 2002) variability in their axes of orientation. This dis-
and the P300 when attending to specified tones organization in the normally uniform alignment of
(Turetsky et al., 1998a, b, 2000a; O’Donnell et al., neurons was interpreted as a developmentally based
1999). The ERP abnormalities suggest dysfunction of disturbance of neuronal migration. However, several
the thalamus and hippocampus in the case of P50, the attempts to replicate these findings with similar and
superior temporal gyrus and perhaps hippocampus different methods have been unsuccessful (Altshuler
and prefrontal cortex in the case of N200 mismatch et al., 1987; Christison et al., 1989; Benes et al., 1991;
negativity, and inferior parietal cortex and probably Arnold et al., 1995; Zaidel et al., 1997).
superior temporal gyrus, thalamus, and hippocampus Abnormal neuronal migration during brain devel-
in the case of P300. opment also was deduced from cytoarchitectural
studies of the entorhinal cortex, a limbic periallo-
cortex intimately related to the hippocampus (Witter
Neuropathological evidence indicating et al., 2000). Jakob and Beckmann (1986, 1989, 1994)
abnormal neurodevelopment and then Arnold et al. (1991a) reported disturbed
cytoarchitectural differentiation, particularly in
While clinical and epidemiological findings present a superficial layers of the entorhinal cortex in schizo-
compelling case for the role of abnormal neurodevel- phrenia. However, others have reported no obvious
opment in the etiology of schizophrenia, characteriz- cytoarchitectural abnormalities in the entorhinal
ing the cellular and molecular mechanism(s) by which cortex (Akil and Lewis, 1997; Krimer et al., 1997).
this might occur continues to present great Quantitative cytoarchitectural studies have attempted
challenges. Neurons in the primate brain are born, to avoid potential subjective bias inherent in
migrate, and assume their mature phenotype in a qualitative cytoarchitectural assessment, but these
complex and highly orchestrated process during too are conflicting. Falkai et al. (1988) found that
fetal development and early childhood period total neuron number in the entorhinal cortex was
(Nowakowski and Rakic, 1981; Arnold and reduced and there was evidence for abnormal clusters
Trojanowski, 1996a, b; Hatten, 1999). By the time of layer II neurons, but Krimer et al. (1997) reported
the clinical expression of schizophrenia is apparent, no alteration in neuronal numbers, density, or layer
usually in the second or third decades of life, volumes in any entorhinal sector, though they did
neurogenesis and much of development is essentially find a trend toward lower numbers and density in
complete. While new neurons are generated in the almost all layers of the most rostral entorhinal
adult brain in certain areas (e.g., the dentate gyrus: sectors. Arnold et al. (1997) applied spatial point
(Eriksson et al., 1998)), their number is small and pattern analysis to interneuronal distances and found
their similarity to perinatal neurons or their role in subtle, but statistically significant differences in
neuroplasticity is still being ascertained (Eisch, 2002). clustering and dispersion indices of neuron position
Neuropathologists have approached the challenge in the entorhinal cortex. Similar approaches in other
of detecting neurodevelopmental anomalies in adult corticolimbic regions (e.g., anterior cingulate cortex)
brains in several ways. Some have focused on have revealed subtle cytoarchitectural differences
324

in schizophrenia compared to controls (Benes and Although aberrant neuronal migration has been
Bird, 1987; Benes et al., 1987, 1992a, 1992b, 1995; the interpretation for most commonly invoked
Chana et al., 2003). Apparent loss of neuropil in the cytoarchitectural abnormalities, there are other
absence of consistent neuronal loss suggests atrophy possible explanations. The ultimate cytoarchitecture
or loss of synaptic connections between neurons of a given region is determined by complex
(Talbot and Arnold, 2002). That is consistent with developmental and regressive neurobiological pro-
increasing indications that synaptic anomalies are cesses. Many changes occur after migration. A host
among the most common abnormalities in schizo- of intrinsic signaling mechanisms responding to
phrenia (Benes, 2000; Mirnics et al., 2000) (Fig. 1). extrinsic growth factors and neurotransmitters
Another cytoarchitectural approach has been to influence neuron survival, morphology, neurite out-
map the number and position of interstitial white growth, and patterns of synapse formation. Those
matter neurons lying deep to cerebrocortical gray young neurons that fail to establish adequate
matter. These neurons are considered to be remnants synaptic connectivity and access to trophic factors
of the cortical subplate and are thought to indicate do not survive. Furthermore, the spatial organization
incomplete neuronal migration during brain develop- of neurons depends not only on the number of
ment. Furthermore, because the subplate is important neurons that migrate to a particular position and
in directing the establishment of normal connectivity, survive, but also the neuropil space in which those
disturbance of the subplate could lead to altered neurons reside. Increases or decreases in this space
formation of connections. Several groups of investi- due to its many cellular (e.g., dendritic, axonal,
gators reported abnormal numbers or positions of glial processes) and extracellular matrix constituents
neurons positive for NADPH-diaphorase or micro- will affect the spatial distribution of neurons. This
tubule-associated protein MAP2 within white matter has been the interpretation of reports of reduc-
of frontal, temporal, and parahippocampal cortices in tions in neuropil in the dorsolateral prefrontal
schizophrenia, although the specific parameters of cortex in schizophrenia (Selemon et al., 1998).
maldistribution varied among studies (Akbarian et al., Finally, environmental factors during growth and
1993, 1996; Anderson et al., 1996; Kirkpatrick et al., development (e.g., infection, ischemic injury,
1999; Rioux et al., 2003). trauma, etc.) as well as neurodegenerative changes

Fig. 1. Cytoarchitectural appearance of the entorhinal cortex from comparable rostral portions, with (a) nonpsychiatric control, and
(b) and (c) two schizophrenia subjects. Compared to the control, the entorhinal cortex in the schizophrenia subjects show subtle
alterations in the cellular composition and spatial arrangement of neurons in layers II and III. Note the poorly formed or absent
clusters of neurons in layer II and the more patchy distribution of neurons in layer III. The consistency of such findings across studies
has been controversial. Adapted from Arnold et al. (1997).
325

that are part of normal aging may further alter the guidance, and synaptogenesis (Burden, 2000; Hall
cytoarchitecture that is observed at postmortem et al., 2000; Coyle-Rink et al., 2002; Castelo-Branco
examination. et al., 2003; Salinas, 2003). Investigations of the Wnt
Investigators have examined the expression of signaling pathway in schizophrenia have produced
various proteins in adult postmortem brain tissues inconclusive findings so far, including reductions in
that play critical roles in brain development. Among b- and g-catenins in the hippocampus (Cotter et al.,
those proteins is reelin, which plays an important role 1998), increased numbers of Wnt-1 immuno-
in neuronal migration and maturation (Tissir and reactive neurons in the hippocampus (Miyaoka
Goffinet, 2003). Decreased reelin-positive cell num- et al., 1999), and decreased GSK3b, but normal
bers and lower reelin mRNA and protein expression b-catenin in prefrontal cortex (Beasley et al., 2001)
have been found by several research groups in while another study in a different cohort by the same
prefrontal cortex, hippocampus, and cerebellum authors found no abnormalities in GSK3b (Beasley
(Impagnatiello et al., 1998; Guidotti et al., 2000; et al., 2002).
Fatemi et al., 2001; Eastwood and Harrison, 2003; Despite the early, conflicting findings, GSK-3 is
Eastwood et al., 2003). During embryogenesis, reelin emerging as a candidate of great interest in
is an extracellular matrix protein secreted primarily schizophrenia with respect to both neurodevelopment
by Cajal–Retzius cells of the cortex that serves to and neuroplasticity (Kozlovsky et al., 2002). Aside
guide neuronal migration (Tissir and Goffinet, 2003), from its role in the Wnt signaling cascade, GSK-3a
stimulate dendritic spine maturation (Liu et al., 2001; and GSK-3b are at a nexus of other signaling
Pappas et al., 2001), and regulate synaptogenesis pathways, including the mitogen-activated protein
(Sarnat and Flores-Sarnat, 2002). Murine or human kinase (MAPK) and phosphatidyl-3 kinase (PI-3K)
reelin mutations severely disrupt brain develop- pathways (Frame and Cohen, 2001; Doble and
ment and result in lissencephaly, inverted cortical Woodgett, 2003). Downstream, it has critical roles
lamination, abnormal axonal connectivity and in the phosphorylation of neural cell adhesion
cerebellar hypoplasia (Pilz et al., 2002), which in molecule, synapsin-1, microtubule-associated protein
humans causes mental retardation, epilepsy, and 1B (MAP1B), MAP2, CREB, the glucocorticoid
neurogenic hypotonia. In the adult, reelin continues receptor, and p53, which are variously involved in
to be expressed primarily in GABAergic local neuronal migration, axon outgrowth and guidance,
circuit neurons of the cortex and hippocampus dendrogenesis and arborization, spine formation and
(Pesold et al., 1998). The function of reelin in the remodeling, synaptogenesis and synapse functioning,
adult nervous system is not known, but its and programmed cell death. Decreases in GSK-3b
preferential expression in apposition to dendritic protein levels and enzyme activity have been reported
spines suggests a possible role in dendritic plasticity in schizophrenia in prefrontal cortex in two separate
(Costa et al., 2001). brain collections (Kozlovsky et al., 2000; Beasley
The Wnt protein/gene family is another example et al., 2001; Kozlovsky et al., 2001) and we have
of a development-related protein that has been observed a significant decrease in the proportion of
evaluated in schizophrenia. While the Wnt signaling GSK-3b immunoreactive neurons in the hippocam-
pathway is complex (Cox and Peifer, 1998), extra- pus in our own collection (unpublished data). As
cellularly secreted Wnt binds to frizzled, a cell elaborated below, the GSK-3 pathway has addition-
membrane receptor that stimulates a complex ally been invoked in recent genetic association studies
signaling cascade, ultimately suppressing the of schizophrenia.
constitutive activity of glycogen synthase kinase 3b
(GSK-3b). This prevents degradation of b-catenin
and leads to increased b-catenin-mediated gene Neuropathological evidence indicating
transcription of important developmentally regula- abnormal synapses
tory genes. Wnt plays critical roles in many phases of
brain development, including neurogenesis, basic It has been theorized that the bizarre thought
patterning of the nervous system, axogenesis, axon processes and cognitive dysfunction of schizophrenia
326

may result from ‘‘miswiring’’ or aberrant patterns or The most commonly used method to assess
integrity of synaptic connections in the brain. Also, synaptic integrity in schizophrenia has been immuno-
neuropsychological studies focused on learning and chemistry of synapse-related proteins with Western
memory and electrophysiological studies have blotting, ELISA, or immunohistochemistry. Most
spawned notions of impaired plasticity and a studies have focused on presynaptic proteins. Among
dysregulation of dynamic interactions within and the proteins that have been examined are the synaptic
across brain systems (Frith and Done, 1988; Gray vesicle membrane, docking, and fusion proteins
et al., 1991; Friston, 1999; Breakspear et al., 2003). synaptophysin, synapsin, SNAP25, complexins I
Synapses have been investigated by a variety of and II, rab3a, and syntaxin, as well as GAP43 and
means in postmortem tissues in schizophrenia. NCAMs which are also highly expressed in axon
Ultrastructural studies have been rare due to the terminals. The regions most commonly examined
technical confounds inherent in human neuropathol- have been hippocampus, dorsolateral prefrontal
ogy research including agonal state and postmortem cortex, and anterior cingulate. While not without
delay effects on the physical integrity of the synaptic controversy, most studies reported significant
complex. Nonetheless, while all ultrastructural find- decreases in presynaptic terminal markers (for
ings await replication, abnormalities have been reviews, see Honer et al., 2000; Eastwood and
reported in the densities and aggregation of synapses Harrison, 2001; Harrison and Eastwood, 2001).
(Soustek, 1989; Aganova and Uranova, 1992; Kung Complementing the immunochemical studies
et al., 1998), dendritic spine morphology and have been studies of presynaptic protein gene
morphometry (Roberts et al., 1996; Kolomeets and expression with Northern, in situ hybridization,
Uranova, 1999), axon terminal mitochondria RT-PCR and microarray analyses. Here too,
(Uranova et al., 1996; Kolomeets and Uranova, significant abnormalities of mRNA expression,
1999), and various other changes in axospinous mostly decreases, have been reported (Honer et al.,
densities and morphologies (for review, see Honer 2000; Eastwood and Harrison, 2001; Harrison and
et al., 2000). Eastwood, 2001).
Abnormalities in dendritic spines and arborization Among the most interesting recent studies have
also have been reported using Golgi impregnation been the gene expression microarray experiments.
and immunohistochemical methods. Golgi studies These have the capacity to examine the coordinated
have described decreased spine densities in prefrontal expression of large numbers of genes and analyze
and temporal cortices and in the hippocampus them by functional groups and pathways. The first
(Glantz and Lewis, 1995; Lewis and Glantz, 1997; published microarray analysis of schizophrenia
Garey et al., 1998; Rosoklija et al., 2000), decreased Mirnics et al. (2000) reported a general down-
densities of pyramidal basilar dendrites in prefrontal regulation of gene transcripts encoding proteins
cortex pyramidal neurons (Broadbelt et al., 2002) and involved in presynaptic function in the dorsolateral
increased densities of dentate gyrus granule cell prefrontal cortex in schizophrenia. The genes that
basilar and recurrent dendrites (Lauer et al., 2003). were most consistently downregulated were N-ethyl-
Another index of dendritic densities that has been malemide sensitive factor (NSF), synapsin II,
used in schizophrenia is immunolabeling for the synaptojanin I and synaptotagmin. In addition,
microtubule-associated protein MAP2 which is various genes involved in GABAergic and glutama-
selectively expressed in the somatodendritic domain tergic neurotransmission as well as a potentially
of neurons. Two studies reported a decrease in the important signal transduction molecule (regulator of
density of MAP2 expression in the hippocampus G protein signaling 4 (RGS4)), and several G0
(Arnold et al., 1991b; Rosoklija et al., 1995) while subunits were decreased in this cohort (Mirnics et al.,
another group found significant increases in MAP2 2001). Hakak et al. (2001) used large oligonucleotide
immunoreactive dendritic arborization in the hippo- microarrays to examine dorsolateral prefrontal cortex
campus (Cotter et al., 2000). Future studies will need in an elderly, highly chronic schizophrenia cohort.
to clarify the nature and extent of dendritic While emphasizing abnormally decreased expression
abnormalities in schizophrenia. of oligodendroglial genes (which also could
327

importantly affect neural connectivity), they also


reported abnormalities in various presynaptic,
GABA, and postsynaptic signal transduction path-
way molecules, albeit with some findings in opposite
directions to those reported by Mirnics et al. The
bases for these discrepancies remain to be elucidated
and may be related to methodological differences in
samples, analytic strategies, or biological variability.
In another microarray study examining a more limi-
ted number of genes in pooled samples of prefrontal
cortex, cerebellum and midtemporal gyrus, Vawter
et al. (2001)reported significant decreases in a number
of genes related to synaptic signaling.
The three studies cited above examined gene
expression in tissue homogenates. In a more novel
approach, Hemby et al. (2002) used single cell
microdissection and antisense RNA amplification
with microarrays to generate an expression profile of
individual entorhinal cortex neurons (Fig. 2). After
immunohistochemical and morphological identifica-
tion in paraffin-embedded sections from elderly, Fig. 2. (a–b) Magnified portions of human gene discovery
microarray images (GDA; Genome Systems, Inc., St. Louis,
chronic subjects with schizophrenia and matched
MO, USA) after hybridization with control (a) and schizo-
controls, mRNA from individually microdissected phrenia (b) mRNA samples amplified from single entorhinal
stellate neurons in layer II of the entorhinal cortex was neurons. Gray arrows indicate site of SNAP-25, the hybridiza-
extracted and linearly amplified. The relative abun- tion intensity of which was decreased by 4.4 fold in
dance of mRNAs so obtained was determined using schizophrenia compared to control. (c) Graph depicts expres-
several cDNA microchip arrays. Over 25,000 genes sion levels using confirmatory reverse Northern blot analysis in
layer II–III entorhinal cortical neurons in control (black bars)
were assessed. Forced clustering of the data revealed and schizophrenia (gray bars) specimens for selected genes
marked differences between schizophrenia subjects including glutamate receptors, G-proteins, and synapse-related
and controls in mRNA expression of presynaptic proteins. Messenger RNA values correspond to hybridization
proteins (syntaxin, synaptotagmins, synaptic vesicle intensity for individual transcripts normalized to the total blot
hybridization intensity. Bars represent mean  SEM. Asterisks
amine transporter, SNAP-25, and g-adaptin), recep-
indicate p < 0.05. Adapted from Hemby et al. (2002).
tors (GABA-A, a-7 nicotinic, interleukin-2, retinoic
acid), ion channels (Naþ, Kþ, Cl), and transcription
factors (e.g., fos, junB, PML-1, Brg, and Brm). by Kim et al. (1980) who noted decreased levels of
While many of the pre- and postsynaptic cerebrospinal fluid glutamate in schizophrenia, the
structural, protein and gene expression findings glutamate hypothesis of schizophrenia has garnered
require clarification and replication, all together, increasing support over the years (for reviews, see
neuropathological evidence provide substantial Grant, 2003; Konradi and Heckers, 2003). This
support for a central role of synaptic abnormalities hypothesis holds that hypofunctioning of the
in schizophrenia. glutamate system, and in particular, NMDA recep-
tors (with secondary increases in glutamate release
in some areas) gives rise to the positive, negative,
Neuropathological evidence indicating abnormal cognitive, and deteriorative features of schizophrenia
synaptic plasticity (Olney et al., 1999).
Apart from its important roles in synaptogenesis
Extensive data implicates dysregulation of the and synaptic toxicity/degeneration, it is well estab-
glutamate system in schizophrenia. First presented lished that one of the most important forms of
328

synaptic plasticity in higher cognition, long term somewhat weaker data for regions of 6q, 10p, 15q,
potentiation (LTP), is NMDA receptor mediated. 16q, 17q, and 18q.
Activation of NMDA receptors leads to Caþþ influx, In the last two years, associations between
activating Caþþ-dependent enzymes such as schizophrenia and a number of specific genes have
CaMKII, which translocates to the nucleus to been reported (for review, see Harrison and Owen,
phosphorylate the transcription factor CREB, 2003 and O’Donovan et al., 2003. While some of
which leads to new protein synthesis important for these have been replicated, others remain unrepli-
synaptic remodeling. Transgenic and knock-out cated or controversial. Specific candidate genes for
experiments in various species have demonstrated schizophrenia now include those encoding, neuregu-
the necessary role for CREB in the establishment lin 1 (NRG1) (Stefansson et al., 2002, 2003a; Tang
of long-term memory via LTP. et al., 2003a; Williams et al., 2003b; Yang et al.,
The NMDA receptor itself is just one component 2003), dysbindin (i.e., dystrobrevin binding protein 1,
of a complex multiprotein postsynaptic machinery DTNBP1) (Straub et al., 2002; Schwab et al., 2003)
that mediates neurotransmission and plasticity. DISC-1 (Millar et al., 2000; Blackwood et al., 2001;
Apart from the NMDA receptor, important compo- Devon et al., 2001; Millar et al., 2001; Hwu et al.,
nents of the postsynaptic density include the 2003), proline dehydrogenase (Jacquet et al., 2002;
scaffolding proteins PSD95, Shank, Homer, Liu et al., 2002; Fan et al., 2003; Williams et al.,
GKAP, GRIP as well as AMPA and metabotropic 2003a), G72/G30 (Chumakov et al., 2002; Hattori
glutamate receptors (Sheng and Kim, 2000). et al., 2003), regulator of G-protein signaling-4
Postmortem studies have reported schizophrenia- (RGS4) (Mirnics et al., 2001; Chowdari et al., 2002),
related abnormalities in the expression of a number the D3 dopamine receptor (Jonsson et al., 2003),
of NMDA receptor complex and related signal D-amino acid oxidase (Chumakov et al., 2002),
transduction proteins, including the NR1, NR2A, catechol-O-methyl transferase (O’Donovan et al.,
and NR28 receptor subunits, PSD95, SAP97, 2003), and AKT (Emamian et al., 2004). While
SAP98, mGlur5, mGluR6, Erk1, MAPK phospha- variations in each of these genes confers only modest
tase, and the excitatory amino acid transporter increases in risk for the disorder, their discovery
EAAT2 (Grant, 2003). Altogether these findings nonetheless identifies candidate proteins and mole-
indicate substantial differences in the molecular cular pathways that may importantly contribute to
machinery of glutamate neurotransmission and the pathophysiology of schizophrenia. This opens the
signal transduction and are consistent with a model door to a functional genomics of schizophrenia. It is
of impaired neuroplasticity in schizophrenia leading noteworthy that most of these genes play important
to the learning and other cognitive deficits typical of roles in neurodevelopment, neurotransmission, or
the disorder. neuroplasticity.
The data supporting the associations of
neuregulin-1, dysbindin, and AKT are among the
strongest. In the ensuing paragraphs, we will focus on
Genes recently associated with schizophrenia: these three genes in schizophrenia and their functions
implications for neurodevelopment and in neurodevelopment, functional synaptic connectiv-
neuroplasticity ity, and neuroplasticity.

Genetic susceptibility plays an important role in the


pathogenesis of schizophrenia, the hereditability of Neuregulin 1 (NRG1)
which is estimated to be 82–84% (Owen et al., 2002).
In a meta-analysis of whole genome scans, Lewis et al. The first report on the linkage and association of
(2003) reported greater consistency of linkage results NRG1 to schizophrenia came from the studies of the
across studies than had previously been recognized deCode Genetics group in the Icelandic population
and identified major susceptibility loci on chromo- (Stefansson et al., 2002). In a genome wide scan, the
somes 1q, 3p, 5q, 6p, 8p, 11q, 14p, 20q, and 22q, with authors found the highest multipoint LOD score of
329

greater than 3 on chromosome 8p, in a region addition, NRG1 modulates migration of neuronal
previously identified by other groups too (Lewis et al., precursors as well as neuronal cells (Schmid et al.,
2003). Extensive fine mapping of the 8p locus and 2003) and thus can provide a lead, considering that
haplotype association analysis as well as the abnormal migration of neurons has been implicated
transmission/disequilibrium test identified a highly for disturbed cytoarchitecture.
significant core haplotype consisting of five single Another important role of NRG1 in the CNS is
nucleotide polymorphisms (SNPs) and two micro- in the development and survival of glial cells. In
satellite markers at the 50 end of the NRG1 gene transgenic mice with null mutations for erbB3 or
associated with schizophrenia. Since the initial report, erbB2, receptors for NRG1, as well as in animals with
NRG1’s association with schizophrenia has been cre based conditional ablation of erbB2, there is a
replicated in several other populations, although no dramatic lack of Schwann cells (Riethmacher et al.,
functional mutations have yet been defined in the 1997; Morris et al., 1999; Woldeyesus et al., 1999).
gene (Stefansson et al., 2002, 2003a, b, 2004; Williams In cultured multipotent neural crest cells, NRG1
et al., 2003b; Yang et al., 2003; Iwata et al., 2004; promotes the adoption of glial cell fate and
Tang et al., 2004). suppresses neuronal differentiation (Shah et al.,
NRG1 has diverse functions in the CNS and thus 1994; Shah and Anderson, 1997). NRG1 is also
the sequence variations in NRG1 may have various required for the development of oligodendroglial
implications in relation to the pathophysiology of lineage. Recently, the density of oligodendrocytes
schizophrenia (Buonanno and Fischbach, 2001). The has been found to decrease in certain regions of post-
effects of NRG1 on N-methyl D-aspartate (NMDA) mortem brains of patients with schizophrenia (Hof
receptors should be seen as a possible link to the et al., 2002, 2003), which is in part corroborated in
pathophysiology of schizophrenia, particularly in microarray analyses (Hakak et al., 2001). Therefore,
light of the glutamatergic hypothesis of schizophrenia it is possible that changes in NRG1 expression
(Konradi and Heckers, 2003). When NRG1 is contribute to oligodendrocyte abnormalities asso-
applied to cerebellar slices, NMDA receptor subunit ciated with schizophrenia.
2C (NR2C) mRNAs increase 100-fold in granule The nervous system is not just comprised of
cells, suggesting that NRG1-erbB signaling directly neuron-neuron connections but also of robust cross
regulates glutamatergic neurotransmission (Ozaki talks between neurons and glial cells (Haydon, 2001).
et al., 1997, 2000). In addition, NRG1 hypomorphic Trophic signaling between neurons and glia is
mice exhibited abnormal prepulse inhibition of the reciprocal and NRG1, released from both neurons
acoustic startle responses which is widely seen as an and glial cells, might play a key role in regulating
endophenotype of schizophrenia. Interestingly, their interactions (Lemke, 2001). Thus, alterations in
clozapine treatment reduced hyperactive behaviors, NRG1 gene, such as in persons with the NRG1 high
as it decreases the binding activities of NMDA risk haplotype, may not only result in the compro-
receptors. Altogether, this evidence suggests a mised availability of NRG1 for trophic support for
possible association between alterations in NRG1- neurons or glial cells, but may also alter interactive
erbB signaling with the changes in glutamatergic signaling between these cells, leading to alterations
neurotransmission observable in schizophrenia. in their synaptic strength (Moises et al., 2002;
NRG1 also affects neuronal development, migra- Falls, 2003).
tion, and survival. When NRG1 is blocked by
ribozyme-tRNA transgene technique in chick
embryos, the proliferation of neuroepithelial cells Dysbindin-1
and the differentiation of retinal ganglion neurons are
found to decrease (Bermingham-McDonogh et al., Dysbindin-1 is a 352 amino acid protein with a
1996; Zhao and Lemke, 1998). In vitro, NRG1 has predicted molecular mass of 40 kDa (Benson et al.,
been shown to support neurite outgrowth in various 2001). It contains a coiled-coil domain between
cell lines (Vaskovsky et al., 2000), and also increase amino acids 88–177 believed to be the binding site
neuronal survival and decrease apoptosis. In of dystrobrevins, the muscle protein myospryn
330

(Benson et al., 2003) and the proteins muted and reduced by as much as 20% in about 40% of
pallidin (Li et al., 2003b), which are important in schizophrenia cases compared to nonpsychiatric
trafficking proteins to lysosome-related organelles. controls (McClintock et al., 2003). Immunohisto-
Dysbindin-1 is encoded by a gene at chromosome chemical studies in the hippocampal formation found
locus 6p22.3 in the human genome (Straub et al., that dysbindin-1 protein levels were significantly
2002). The dysbindin-1 gene and its protein are reduced by as much as 42% in one or more
widely expressed in the body, including the brain presynaptic fields of intrinsic glutamatergic connec-
(Benson et al., 2001; Talbot et al., 2004). The protein tions in 93% of schizophrenia cases compared to
is found in neuronal cell bodies of many brain areas nonpsychiatric controls matched for age, sex,
and is markedly enriched in a few presynaptic fields, cerebral hemisphere, type of fixative, and postmor-
specifically those of the hippocampal formation, tem interval (Talbot et al., 2004). The findings in the
striatum, substantia nigra, deep cerebellar nuclei, dentate portion of the hippocampal formation were
inferior olive, and substantia gelatinosa of the spinal replicated in an independent sample. No such
cord (Arnold et al., 2003). differences were found in bipolar or major depressive
Interest in the protein escalated with the 2002 comparison cases, though the latter showed a trend in
report that certain haplotypes of SNPs in the that direction. Morphometric analyses on our cases
dysbindin-1 gene are significantly associated with revealed that the observed presynaptic dysbindin-1
schizophrenia (Straub et al., 2002). Within two years reductions in the hippocampal formation were not
of that discovery, the association of schizophrenia due to axon terminal loss. Nor were the reductions
with one or another dysbindin-1 haplotype (com- readily attributed to neuroleptic effects, because
posed of 3–8 SNPs) has been confirmed in large control tests on mice treated chronically with
populations from England (Datta et al., 2003; van haloperidol at therapeutic doses showed no effect
den Oord et al., 2003), Ireland (van den Oord et al., on immunoreactivity of dysbindin-1 in the hippo-
2003; Williams et al., 2003b), Wales (Williams et al., campal formation (Fig. 3).
2003b), the Netherlands (Bakker et al., 2003), The prefrontal cortex and hippocampal formation
Germany/Hungary/Israel (Schwab et al., 2003), expression studies suggest that dysbindin-1 reduc-
Sweden (Van Den Bogaert et al., 2003), China (Shi tions in the brain may be a common feature in
et al., 2003; Tang et al., 2003), and Japan (Yamada schizophrenia. Since the percentage of cases showing
et al., 2003). Only two studies failed to replicate such such reductions in the hippocampal formation was
findings. One analyzed an Irish population (Morris far higher than reported frequencies of dysbindin-1
et al., 2003) in which subsequent analysis showed haplotypes conferring elevated risk of schizophrenia
that a different dysbindin haplotype (with a SNP in (Straub et al., 2002; Van Den Bogaert et al., 2003;
the promoter region) actually was more common in van den Oord et al., 2003), it seems likely that those
schizophrenia than in control cases (O’Donovan haplotypes are not the only factor contributing to the
et al., 2003; Williams et al., 2003c). The remaining reduced protein levels. That suggests important roles
negative study on German and Polish populations for other genetic, as well as nongenetic factors
did not test haplotypes including the promoter SNP unknown at present. Among them may be neuro-
(Van Den Bogaert et al., 2003). No other gene to date developmental factors affecting protein expression,
has been associated with schizophrenia so frequently posttranslational modification, transport, and/or
and consistently. degradation. Dysbindin-1 reductions may simply be
Such findings led O’Donovan and his colleagues the common outcome of many factors increasing risk
(O’Donovan et al., 2003) to argue that the dysbindin- of schizophrenia, whether they are genetic, neuro-
1 gene is among the most promising genetic developmental, or environmental.
susceptibility locus for schizophrenia. Indeed, two Our studies of dysbindin-1 reductions in the
groups recently reported abnormal dysbindin-1 hippocampal formation point to a role for the protein
protein expression in schizophrenia. Homogenate in regulating glutamatergic transmission. As noted
studies on the prefrontal cortex found that dysbindin- above, we found significant reductions of dysbindin-1
1 gene and protein expression were significantly in intrinsic glutamatergic connections throughout
331

Fig. 3. Dysbindin-1 immunohistochemistry in human hippocampal formation with obvious decrease in dysbindin-1 expression in
many hippocampal sectors in the schizophrenia compared to its matched control. Higher magnifications on right of boxed region of
dentate gyrus shows severe deficit in schizophrenia case of dysbinin-1 protein expression in the granule cells and most notably the inner
molecular layer, an intrinsic glutamatergic terminal field emanating from mossy neurons of the hilus of the dentate gyrus. Adapted
from Talbot et al. (2004).

the hippocampal formation in schizophrenia, speci- deletion mutation in the dysbindin-1 gene causing
fically in hippocampal fields CA1-3, the subiculum, markedly reduced brain levels of dysbindin-1 (Li et al.,
and especially the dentate gyrus (Talbot et al., 2004). 2003b). Consistent with our hypothesis, preliminary
In the core or hilus of the dentate gyrus are large results indicate that sandy mice with the lowest levels
neurons intensely immunoreactive for dysbindin-1. of dysbindin-1 (homozygous animals) have higher
They innervate the inner molecular layer of the VGluT-1 levels in the dentate gyrus than sandy
dentate gyrus, where dysbindin-1 and vesicular mice with higher dysbindin-1 levels (heterozygous
glutamate transporter-1 (VGluT-1) are coextensively animals).
localized in axon terminals. That band of axon Altered glutamatergic neurotransmission in the
terminals displayed a reduction in dysbindin-1 and a hippocampal formation can alter information flow
significantly correlated increase in VGluT-1 in our and neuroplasticity in that structure. Via their
schizophrenia cases compared to matched controls. It glutamatergic output to the inner molecular layer of
is possible then, that dysbindin-1 normally inhibits the dentate gyrus, mossy cells positive for dysbindin-1
glutamate transport into synaptic vesicles and in the hilus of the gyrus can prime or recruit large
thereby regulates glutamatergic transmission in the numbers of granule cells to respond to entorhinal
hippocampal formation. To test that hypothesis, we input (Jackson and Scharfman, 1996). Such cells do
have begun studies on sandy mice, which have a so in a manner found to support long-term
332

potentiation in granule cells of the dentate gyrus rodents that had been chronically treated with
(Hetherington et al., 1994), which project promi- haloperidol.
nently to hippocampal field CA3. The pyramidal cells To further characterize AKT1’s role in schizo-
positive for dysbindin-1 in CA3 are also gluta- phrenia, the authors then examined whether certain
matergic and give rise to collateralized axons, some of variants of the gene encoding AKT1 were preferen-
which terminate locally. Such recurrent collaterals tially transmitted. They genotyped five SNPs at the
provide an excitatory feedback pathway in CA3, AKT1 locus in 268 affected families and identified
impairment of which may contribute to the associa- one 3 SNP haplotype that was preferentially
tive memory deficits observed in mice with a localized transmitted to probands with schizophrenia with a
deficit of NMDA receptors in CA3 (Nakazawa et al., ratio of 1.8:1 ( p ¼ 0.0006), as well as several other
2002). Other collaterals of CA3 pyramidal cells multi-SNP haplotypes that were transmitted at lower
(Schaffer collaterals) project to CA1, where their levels of significance. They then showed that the core
output can induce long-term potentiation via risk haplotype was associated with lower protein
glutamatergic mechanisms (Petrozzino and Connor, levels of AKT1 in lymphocyte cell lines.
1994; Matthies et al., 1995). As with hilar DG As a behavioral and pharmacological assay of
projections, then, reduction of dysbindin-1 observed potential effects of AKT1 deficiency, the authors
in CA3 projections may alter information processing examined prepulse inhibition of the acoustic startle
and diminish synaptic plasticity in the hippocampal response in AKT1 knock-out mice. This is an index
formation. of sensorimotor gating that is abnormal in schizo-
phrenia and which can be modeled in animals. While
there were no baseline differences between AKT1
AKT1 deficient and wild-type mice for startle amplitude
or prepulse inhibition, the knock-out mice exhibi-
AKT1 (v-akt murine thymoma viral oncogene ted reduced prepulse inhibition when additionally
homolog 1), also known as protein kinase B, is one challenged with amphetamine, suggesting some impair-
of the most recently reported genes to be associated ment in the integrity or pharmacological respon-
with schizophrenia (Emamian et al., 2004). In siveness of neural circuitry of sensorimotor gating.
contrast to NRG1 or dysbindin-1 which were first AKT1 is a serine-threonine kinase that is a nexus
identified by fine mapping of known susceptibility for many signal transduction pathways. It plays a
loci, the association of AKT1 with schizophrenia central role in many cellular processes including cell
was identified while examining general hypotheses proliferation, apoptosis, glucose metabolism, angio-
that protein kinases are altered in psychiatric disease. genesis, and cell motility (Brazil et al., 2002). AKT
In particular, it was found that important mood- is activated rapidly by extracellular factors such as
stabilizing psychotropic medications share a mecha- insulin and insulin-like growth factors, and intracel-
nism of action that is dependent upon GSK-3, which lular signals such as Ras. During embryogenesis,
is regulated directly by AKT1 (Coyle and Duman, AKT1 promotes cell proliferation and cell survival
2003). in response to growth factors and stimuli that elicit
Emamian et al. (2004) found that among a panel calcium influx (Dudek et al., 1997) and phosphory-
of kinases involved in synaptic plasticity, AKT1 lates a number of proapoptotic proteins, thereby
protein levels and levels of phosphorylation of one of suppressing death signals (Datta et al., 1999).
its major substrates, GSK-3b were significantly While found in a variety of peripheral tissues,
decreased in lymphocytes from patients with schizo- AKT is highly expressed in the central nervous
phrenia as well as in postmortem prefrontal and system. During embryogenesis it is required for axon
hippocampal brain tissues from two independent elongation and branching (Markus et al., 2002) and
schizophrenia brain tissue collections. GSK3b plays a role in reelin-mediated migration of nascent
protein levels were unaltered. AKT1 and GSK3b neurons to the cortex (Beffert et al., 2002). AKT
phosphorylation did not correlate with age, gender, also is important in regulating metabolic path-
or postmortem interval and were not altered in ways important for cell survival through the
333

neuregulin-mediated PI3 kinase signaling pathway First, schizophrenia is clinically heterogeneous


(Li et al., 2003a). That AKT is important in and almost certainly etiologically heterogeneous,
regulating two other candidate molecules (reelin resulting from interaction of what are probably
and neuregulin) in schizophrenia raises the possibility multiple genetic factors (each of variable effect) and
that multiple genes, each of small effect, may interact multiple environmental factors (each also of variable
to promote the development of schizophrenia. effect). Different combinations of these factors may
AKT1 continues to play important roles in the culminate in the phenotype we classify as schizo-
plasticity of the adult nervous system. Increased phrenia. Investigations to date have had only modest
phosphorylation of AKT has been reported in successes in accommodating heterogeneity in their
response to LTP in the hippocampus and fear design and interpretation of data.
conditioning in the hippocampus (Sanna et al., 2002) Second, schizophrenia typically cannot be diag-
and amygdala in rodents (Lin et al., 2001). AKT1 is nosed until the late teens or early twenties, when most
found in the synaptic plasma membrane and at lower of the fundamental processes of brain development
levels in the dendritic lipid raft and postsynaptic are largely complete. Thus, we must infer anomalous
density fractions of neuronal tissues (Asaki et al., development based on observable features in the
2003). Here it is thought to play a role in a post- adult. For cellular and molecular studies examining
synaptically localized protein synthesis system that the cytoarchitectural appearance and molecular
is one of the mechanisms of synaptic plasticity. composition of the adult brain at the time of autopsy,
Postsynaptic protein synthesis allows local and rapid this occurs at least a few years after most
synthesis of the key proteins required for dendritic developmental events happened, and unless death is
spine or postsynaptic density modifications that unnaturally early (e.g., due to suicide), it usually
occur with long-term memory, the late phase of occurs decades after disease diagnosis.
LTP or long-term depression, and trophic stimula- Etiology is further obscured by the confounding
tion (Kang and Schuman, 1996; Huber et al., 2000; effects of age, medical and psychiatric comorbidity,
Akama and McEwen, 2003). AKT phosphorylates medication and other somatic treatments, environ-
the GABAA receptor that mediates fast inhibitory mental stimulation, and in the case of neuropatho-
synaptic transmission, increasing the density of logical studies, agonal and postmortem influences on
GABAA receptors and thereby increasing synaptic cellular and molecular integrity. To varying degrees,
strength (Wang et al., 2003). Finally, brain-derived these limitations been addressed in the studies
neurotrophic factor signaling phosphorylates AKT reviewed above, and it is critical to remain vigilant
in the hippocampus during spatial reference and about them in interpreting the data.
working memory formation (Mizuno et al., 2003). Finally, to our knowledge, schizophrenia is a
uniquely human disease. Some animal models do
allow us to investigate isolated features of schizo-
Concluding remarks phrenia (i.e., endophenotypes), including neurodevel-
opmental and neuroplastic aspects of these features.
This review has described clinical and neurobiological But such models are partial, and none has yet been
data relevant to the concepts of schizophrenia as a identified or designed to truly encompass a sufficient
disorder of neurodevelopment with consequent range of phenotypic abnormalities to mimic human
effects on connectivity, and ongoing neuroplasticity. schizophrenia.
Exciting findings have emerged from application of Even with these limitations, an increasingly strong
diverse methods to elucidate specific structural case is emerging for schizophrenia as a disease of
abnormalities and identify candidate molecular neurodevelopment, neural connectivity, and neuro-
pathways pertinent to the pathogenesis and patho- plasticity in specific neural and neurochemical path-
physiology of the illness. It is important, however, to ways. Especially noteworthy are converging data
remain mindful of conceptual and practical limita- from genetic association studies, molecular neuro-
tions, which must guide interpretation of the data pathology studies, and selective animal models that
and inform future research strategies. explore the implications of candidate molecules.
334

Identification and confirmation of genetic suscep- Altshuler, L.L., Conrad, A., Kovelman, J.A. and Scheibel, A.
tibility loci (e.g., AKT1, dysbindin, GSK-3, neur- (1987) Hippocampal pyramidal cell orientation in
schizophrenia. Archives of General Psychiatry, 44:
egulin, and reelin) and of molecular mechanism their
1094–1098.
proteins affect (e.g., glutamatergic neurotransmis- Anderson, S.A., Volk, D.W. and Lewis, D.A. (1996) Increased
sion) represent critical steps towards defining the density of microtubule associated protein 2-immunoreactive
neurobiology of schizophrenia and perhaps ulti- neurons in the prefrontal white matter of schizophrenic
mately discovering means of preventing and treating subjects. Schizophrenia Research, 19: 111–119.
it more effectively. Andres, P. (2003) Frontal cortex as the central executive of
working memory: time to revise our view. Cortex, 39:
871–895.
Arnold, S.E., Franz, B.R., Gur, R.C., Gur, R.E.,
Acknowledgments Shapiro, R.M., Moberg, P.J. and Trojanowski, J.Q. (1995)
Smaller neuron size in schizophrenia in hippocampal sub-
Support for our work cited here was provided by the fields that mediate cortical-hippocampal interactions. Am. J.
Psychiatry, 152: 738–748.
National Institutes of Health (MH64045, MH55199,
Arnold, S.E., Han, L.Y., Moberg, P.J., Turetsky, B.I.,
AG10124, AG09215), the National Alliance for Gur, R.E., Trojanowski, J.Q. and Hahn, C.G. (2001)
Research on Schizophrenia and Depression and the Dysregulation of olfactory receptor neuron lineage in
Stanley Foundation. schizophrenia. Arch. Gen. Psychiatry, 58: 829–835.
Arnold, S.E., Hyman, B.T., Van Hoesen, G.W. and
Damasio, A.R. (1991a) Some cytoarchitectural abnormalities
References of the entorhinal cortex in schizophrenia. Arch. Gen.
Psychiatry, 48: 625–632.
Abi-Dargham, A., Mawlawi, O., Lombardo, I., Gil, R., Arnold, S.E., Lee, V.M., Gur, R.E. and Trojanowski, J.Q.
Martinez, D., Huang, Y., Hwang, D.R., Keilp, J., (1991b) Abnormal expression of two microtubule-associated
Kochan, L., Van Heertum, R., Gorman, J.M. and proteins (MAP2 and MAP5) in specific subfields of the
Laruelle, M. (2002) Prefrontal dopamine D1 receptors and hippocampal formation in schizophrenia. Proc. Natl. Acad.
working memory in schizophrenia. J. Neurosci., 22: Sci. USA, 88: 10850–10854.
3708–3719. Arnold, S.E. and Rioux, L. (2001) Challenges, status, and
Abi-Dargham, A. and Moore, H. (2003) Prefrontal DA opportunities for studying developmental neuropathology
transmission at D1 receptors and the pathology of schizo- in adult schizophrenia. Schizophr. Bull., 27: 395–416.
phrenia. Neuroscientist, 9: 404–416. Arnold, S.E., Ruscheinsky, D.D. and Han, L.Y. (1997) Further
Aganova, E.A. and Uranova, N.A. (1992) Morphometric evidence of abnormal cytoarchitecture of the entorhinal
analysis of synaptic contacts in the anterior limbic cortex cortex in schizophrenia using spatial point pattern analyses.
in the endogenous psychoses. Neurosci. Behav. Physiol., 22: Biol. Psychiatry, 42: 639–647.
59–65. Arnold, S.E., Talbot, K., Eidem, W.L., Benson, M.A.,
Akama, K.T. and McEwen, B.S. (2003) Estrogen stimulates Trojanowski, J.Q., Blake, D.J. and Uryu, K. (2003)
postsynaptic density-95 rapid protein synthesis via the Dysbindin distribution in the brain of rodents and humans.
Akt/protein kinase B pathway. J. Neurosci., 23: 2333–2339. Society for Neuroscience, 33: (in press).
Akbarian, S., Kim, J.J., Potkin, S.G., Hetrick, W.P., Arnold, S.E. and Trojanowski, J.Q. (1996a) Human fetal
Bunney, W.E. and Jones, E.G. (1996) Maldistribution of hippocampal development: I. Cytoarchitecture, myeloarchi-
interstitial neurons in prefrontal white matter of the brains of tecture, and neuronal morphologic features. J. Comp.
schizophrenic patients. Archives of General Psychiatry, 53: Neurol., 367: 274–292.
425–436. Arnold, S.E. and Trojanowski, J.Q. (1996b) Human fetal
Akbarian, S., Vinuela, A., Kim, J.J., Potkin, S.G., hippocampal development: II. The neuronal cytoskeleton.
Bunney, W.E. and Jones, E.G. (1993) Distorted distribution J. Comp. Neurol., 367: 293–307.
of nicotinamide-adenine dinucleotide phosphate-diaphorase Arnold, S.E., Trojanowski, J.Q., Gur, R.E., Blackwell, P.,
neurons in temporal lobe of schizophrenics implies anom- Han, L.Y. and Choi, C. (1998) Absence of neurodegeneration
alous cortical development. Archives of General Psychiatry, and neural injury in the cerebral cortex in a sample of elderly
50: 178–187. patients with schizophrenia. Arch. Gen. Psychiatry, 55:
Akil, M. and Lewis, D.A. (1997) Cytoarchitecture of the 225–232.
entorhinal cortex in schizophrenia. American Journal of Asaki, C., Usuda, N., Nakazawa, A., Kametani, K. and
Psychiatry, 154: 1010–1012. Suzuki, T. (2003) Localization of translational components
335

at the ultramicroscopic level at postsynaptic sites of the rat Benson, M.A., Tinsley, C.L. and Blake, D.J. (2003) Myospryn
brain. Brain Res., 972: 168–176. is a novel binding partner for dysbindin in muscle. J. Biol.
Bakker, S.C., Hoogendoom, M.L.C., Sinke, R.J., Chem.
Verzibergen, K.F., Kusters, K.A., Otten, H.G., Pearson, P.L. Bermingham-McDonogh, O., McCabe, K.L. and Reh, T.A.
and Kahn, R.S. (2003) Association study of the neuregulin, (1996) Effects of GGF/neuregulins on neuronal survival and
dysbindin and G72 genes in a large sample of Dutch neurite outgrowth correlate with erbB2/neu expression in
schizophrenic patients. In: Barden N., Cote M., DeLisi L., developing rat retina. Development, 122: 1427–1438.
Gill M., Kelsoe J. and Schalling M. (Eds.), XIth World Blackwood, D.H., Fordyce, A., Walker, M.T., St. Clair, D.M.,
Congress of Psychiatric Genetics, Quebec, 19 p. Porteous, D.J. and Muir, W.J. (2001) Schizophrenia and
Beasley, C., Cotter, D. and Everall, I. (2002) An investigation affective disorders — cosegregation with a translocation at
of the Wnt-signalling pathway in the prefrontal cortex chromosome 1q42 that directly disrupts brain-expressed
in schizophrenia, bipolar disorder and major depressive genes: clinical and P300 findings in a family. Am. J. Hum.
disorder. Schizophr. Res., 58: 63–67. Genet., 69: 428–433.
Beasley, C., Cotter, D., Khan, N., Pollard, C., Sheppard, P., Bleuler, E. (1924) Textbook of Psychiatry. Macmillan,
Varndell, I., Lovestone, S., Anderton, B. and Everall, I. New York.
(2001) Glycogen synthase kinase-3beta immunoreactivity is Braff, D.L., Geyer, M.A., Light, G.A., Sprock, J., Perry, W.,
reduced in the prefrontal cortex in schizophrenia. Neurosci. Cadenhead, K.S. and Swerdlow, N.R. (2001) Impact of
Lett., 302: 117–120. prepulse characteristics on the detection of sensorimotor
Beffert, U., Morfini, G., Bock, H.H., Reyna, H., Brady, S.T. gating deficits in schizophrenia. Schizophr. Res., 49: 171–178.
and Herz, J. (2002) Reelin-mediated signaling locally Braff, D.L., Grillon, C. and Geyer, M.A. (1992) Gating and
regulates protein kinase B/Akt and glycogen synthase kinase habituation of the startle reflex in schizophrenic patients.
3beta. J. Biol. Chem., 277: 49958–49964. Arch. Gen. Psychiatry, 49: 206–215.
Benes, F.M. (2000) Emerging principles of altered neural Brazil, D.P., Park, J. and Hemmings, B.A. (2002) PKB binding
circuitry in schizophrenia. Brain Res. Brain Res. Rev., 31: proteins. Getting in on the Akt. Cell, 111: 293–303.
251–269. Breakspear, M., Terry, J.R., Friston, K.J., Harris, A.W.,
Benes, F.M. and Bird, E.D. (1987) An analysis of the Williams, L.M., Brown, K., Brennan, J. and Gordon, E.
arrangement of neurons in the cingulate cortex of schizo- (2003) A disturbance of nonlinear interdependence in scalp
phrenic patients. Archives of General Psychiatry, 44: EEG of subjects with first episode schizophrenia.
608–616. Neuroimage, 20: 466–478.
Benes, F.M., Majocha, R., Bird, E.D. and Marrotta, C.A. Broadbelt, K., Byne, W. and Jones, L.B. (2002) Evidence for a
(1987) Increased vertical axon numbers in cingulate cortex decrease in basilar dendrites of pyramidal cells in schizo-
of schizophrenics. Archives of General Psychiatry, 44: phrenic medial prefrontal cortex. Schizophr. Res., 58: 75–81.
1017–1021. Brown, A.S., Cohen, P., Harkavy-Friedman, J., Babulas, V.,
Benes, F.M., Sorensen, I. and Bird, E.D. (1991) Reduced Malaspina, D., Gorman, J.M. and Susser, E.S. (2001) A.E.
neuronal size in posterior hippocampus of schizophrenic Bennett Research Award. Prenatal rubella, premorbid
patients. Schizophrenia Bulletin, 17: 597–608. abnormalities, and adult schizophrenia. Biol. Psychiatry,
Benes, F.M., Sorensen, I., Vincent, S.L., Bird, E.D. and 49: 473–486.
Sathi, M. (1992a) Increased density of glutamate-immuno- Brown, A.S. and Susser, E.S. (2002) In utero infection and
reactive vertical processes in superficial laminae in cin- adult schizophrenia. Ment. Retard. Dev. Disabil. Res. Rev.,
gulate cortex of schizophrenic brain. Cerebral Cortex, 2: 8: 51–57.
503–512. Brown, K.J., Gonsalvez, C.J., Harris, A.W., Williams, L.M.
Benes, F.M., Todtenkopf, M.S. and Taylor, J.B. (1995) A shift and Gordon, E. (2002) Target and non-target ERP dis-
in tyrosine hydroxylase-immunoreactive varicosities ITH- turbances in first episode vs. chronic schizophrenia. Clin.
IRv) for pyramidal (PN) to nonpyramidal (NP) neurons Neurophysiol., 113: 1754–1763.
occurs in layer II of the anterior cingulate cortex of Buchsbaum, M.S., Nenadic, I., Hazlett, E.A., Spiegel-Cohen, J.,
schizoprenics. Society for Neuroscience Abstracts, 21: 259. Fleischman, M.B., Akhavan, A., Silverman, J.M. and
Benes, F.M., Vincent, S.L., Alsterberg, G., Bird, E.D. and Siever, L.J. (2002) Differential metabolic rates in prefrontal
SanGiovanni, J.P. (1992b) Increased GABAa receptor and temporal Brodmann areas in schizophrenia and schizo-
binding in superficial layers of cingulate cortex in schizo- typal personality disorder. Schizophr. Res., 54: 141–150.
phrenics. Journal of Neuroscience, 12: 924–929. Buckner, R.L., Logan, J., Donaldson, D.I. and Wheeler, M.E.
Benson, M.A., Newey, S.E., Martin-Rendon, E., Hawkes, R. (2000) Cognitive neuroscience of episodic memory encoding.
and Blake, D.J. (2001) Dysbindin, a novel coiled-coil- Acta Psychol. (Amst.), 105: 127–139.
containing protein that interacts with the dystrobrevins in Buka, S.L., Tsuang, M.T., Torrey, E.F., Klebanoff, M.A.,
muscle and brain. J. Biol. Chem., 276: 24232–24241. Wagner, R.L. and Yolken, R.H. (2001) Maternal cytokine
336

levels during pregnancy and adult psychosis. Brain Behav. Macciardi, F., Sham, P.C., Straub, R.E., Weinberger, D.R.,
Immun., 15: 411–420. Cohen, N., Cohen, D., Ouelette, G. and Realson, J. (2002)
Buonanno, A. and Fischbach, G.D. (2001) Neuregulin and Genetic and physiological data implicating the new
ErbB receptor signaling pathways in the nervous system. human gene G72 and the gene for D-amino acid oxidase
Curr. Opin. Neurobiol., 11: 287–296. in schizophrenia. Proc. Natl. Acad. Sci. USA, 99:
Burden, S.J. (2000) Wnts as retrograde signals for axon and 13675–13680.
growth cone differentiation. Cell, 100: 495–497. Clementz, B.A., Keil, A. and Kissler, J. (2004) Aberrant brain
Cannon, M., Jones, P.B. and Murray, R.M. (2002) Obstetric dynamics in schizophrenia: delayed buildup and prolonged
complications and schizophrenia: historical and meta-analy- decay of the visual steady-state response. Brain Res. Cogn.
tic review. Am. J. Psychiatry, 159: 1080–1092. Brain Res., 18: 121–129.
Cantor-Graae, E., Ismail, B. and McNeil, T.F. (1998) Neonatal Conrad, A.J., Abebe, T., Austin, R., Forsythe, S. and
head circumference and related indices of disturbed fetal Scheibel, A.B. (1991) Hippocampal pyramidal cell disarray
development in schizophrenic patients. Schizophr. Res., 32: in schizophrenia as a bilateral phenomenon. Archives of
191–199. General Psychiatry, 48: 413–417.
Castelo-Branco, G., Wagner, J., Rodriguez, F.J., Kele, J., Costa, E., Davis, J., Grayson, D.R., Guidotti, A., Pappas, G.D.
Sousa, K., Rawal, N., Pasolli, H.A., Fuchs, E., Kitajewski, J. and Pesold, C. (2001) Dendritic spine hypoplasticity and
and Arenas, E. (2003) Differential regulation of midbrain downregulation of reelin and GABAergic tone in schizo-
dopaminergic neuron development by Wnt-1, Wnt-3a, and phrenia vulnerability. Neurobiol. Dis., 8: 723–742.
Wnt-5a. Proc. Natl. Acad. Sci. USA, 100: 12747–12752. Cotter, D., Kerwin, R., al-Sarraji, S., Brion, J.P., Chadwich, A.,
Chana, G., Landau, S., Beasley, C., Everall, I.P. and Cotter, D. Lovestone, S., Anderton, B. and Everall, I. (1998)
(2003) Two-dimensional assessment of cytoarchitecture in Abnormalities of Wnt signalling in schizophrenia — evidence
the anterior cingulate cortex in major depressive disorder, for neurodevelopmental abnormality. Neuroreport, 9:
bipolar disorder, and schizophrenia: evidence for decreased 1379–1383.
neuronal somal size and increased neuronal density. Biol. Cotter, D., Wilson, S., Roberts, E., Kerwin, R. and Everall, I.P.
Psychiatry, 53: 1086–1098. (2000) Increased dendritic MAP2 expression in the hippo-
Chowdari, K.V., Mirnics, K., Semwal, P., Wood, J., campus in schizophrenia. Schizophr. Res., 41: 313–R323.
Lawrence, E., Bhatia, T., Deshpande, S.N., Thelma, B.K., Cox, R.T. and Peifer, M. (1998) Wingless signaling: the
Ferrell, R.E., Middleton, F.A., Devlin, B., Levitt, P., inconvenient complexities of life. Curr. Biol., 8: R140–144.
Lewis, D.A. and Nimgaonkar, V.L. (2002) Association and Coyle, J.T. and Duman, R.S. (2003) Finding the intracellular
linkage analyses of RGS4 polymorphisms in schizophrenia. signaling pathways affected by mood disorder treatments.
Hum. Mol. Genet., 11: 1373–1380. Neuron, 38: 157–160.
Christison, G.W., Casanova, M.F., Weinberger, D.R., Coyle-Rink, J., Del Valle, L., Sweet, T., Khalili, K. and
Rawlings, R. and Kleinman, J.E. (1989) A quantitative Amini, S. (2002) Developmental expression of Wnt signaling
investigation of hippocampal pyramidal cell size, shape, and factors in mouse brain. Cancer. Biol. Ther., 1: 640–645.
variability of orientation in schizophrenia. Archives of Datta, S.R., Brunet, A. and Greenberg, M.E. (1999) Cellular
General Psychiatry, 46: 1027–1032. survival: a play in three Akts. Genes Dev., 13: 2905–2927.
Chua, S.E. and Murray, R.M. (1996) The neurodevelopmental Datta, S.R., Rizig, M.A., McQuillin, A., Kalsi, G.,
theory of schizophrenia: evidence concerning structure and Lawrence, J., Quested, D., Moorey, H., Lamb, G.,
neuropsychology. Ann. Med., 28: 547–555. Chowdhury, U., Chowdhury, V., Krasucki, R., Pimm, J.,
Chumakov, I., Blumenfeld, M., Guerassimenko, O., Curtis, D., Muir, W., Blackwood, D. and Gurling, H.M.D.
Cavarec, L., Palicio, M., Abderrahim, H., Bougueleret, L., (2003) Tests of linkage disequilibrium between schizophrenia
Barry, C., Tanaka, H., La Rosa, P., Puech, A., Tahri, N., and genetic markers at the G72 and dysbindin loci.
Cohen-Akenine, A., Delabrosse, S., Lissarrague, S., In: Barden N., Côté M., DeLisi L., Gill M., Kelsoe J. and
Picard, F.P., Maurice, K., Essioux, L., Millasseau, P., Schalling M. (Eds.), XIth World Congress of Psychiatric
Grel, P., Debailleul, V., Simon, A.M., Caterina, D., Genetics, Quebec, pp. 131–172.
Dufaure, I., Malekzadeh, K., Belova, M., Luan, J.J., Davidson, M., Reichenberg, A., Rabinowitz, J., Weiser, M.,
Bouillot, M., Sambucy, J.L., Primas, G., Saumier, M., Kaplan, Z. and Mark, M. (1999) Behavioral and intellectual
Boubkiri, N., Martin-Saumier, S., Nasroune, M., markers for schizophrenia in apparently healthy male
Peixoto, H., Delaye, A., Pinchot, V., Bastucci, M., adolescents. Am. J. Psychiatry, 156: 1328–1335.
Guillou, S., Chevillon, M., Sainz-Fuertes, R., Meguenni, S., Devon, R.S., Anderson, S., Teague, P.W., Burgess, P.,
Aurich-Costa, J., Cherif, D., Gimalac, A., Van Duijn, C., Kipari, T.M., Semple, C.A., Millar, J.K., Muir, W.J.,
Gauvreau, D., Ouellette, G., Fortier, I., Raelson, J., Murray, V., Pelosi, A.J., Blackwood, D.H. and
Sherbatich, T., Riazanskaia, N., Rogaev, E., Porteous, D.J. (2001) Identification of polymorphisms
Raeymaekers, P., Aerssens, J., Konings, F., Luyten, W., within disrupted in schizophrenia 1 and disrupted in
337

Schizophrenia 2, and an investigation of their association Falkai, P., Bogerts, B. and Rozumek, M. (1988) Limbic
with schizophrenia and bipolar affective disorder. Psychiatr. pathology in schizophrenia: the entorhinal region — a
Genet., 11: 71–78. morphometric study. Biological Psychiatry, 24: 515–521.
Doble, B.W. and Woodgett, J.R. (2003) GSK-3: tricks of the Fallgatter, A.J. (2001) Electrophysiology of the prefrontal
trade for a multi-tasking kinase. J. Cell Sci., 116: 1175–1186. cortex in healthy controls and schizophrenic patients: a
Done, D., Crow, T.J., Johnstone, E. and Sacker, A. (1994) review. J. Neural. Transm., 108: 679–694.
Childhood antecedents of schizophrenia and affective illness: Falls, D.L. (2003) Neuregulins: functions, forms, and signaling
social adjustment at ages 7 and 11. British Medical Journal, strategies. Exp. Cell Res., 284: 14–30.
1–12. Fan, J.B., Ma, J., Zhang, C.S., Tang, J.X., Gu, N.F.,
Dudek, H., Datta, S.R., Franke, T.F., Birnbaum, M.J., Yao, R., Feng, G.Y., St. Clair, D. and He, L. (2003) A family-based
Cooper, G.M., Segal, R.A., Kaplan, D.R. and association study of T1945C polymorphism in the proline
Greenberg, M.E. (1997) Regulation of neuronal survival by dehydrogenase gene and schizophrenia in the Chinese
the serine-threonine protein kinase Akt. Science, 275: population. Neurosci. Lett., 338: 252–254.
661–665. Fatemi, S.H., Kroll, J.L. and Stary, J.M. (2001) Altered levels
Eastwood, S.L. and Harrison, P.J. (2001) Synaptic pathology of Reelin and its isoforms in schizophrenia and mood
in the anterior cingulate cortex in schizophrenia and mood disorders. Neuroreport, 12: 3209–3215.
disorders. A review and a Western blot study of synapto- Faw, B. (2003) Pre-frontal executive committee for perception,
physin, GAP-43 and the complexins. Brain Res. Bull, 55: working memory, attention, long-term memory, motor
569–578. control, and thinking: a tutorial review. Conscious Cogn.,
Eastwood, S.L. and Harrison, P.J. (2003) Interstitial white 12: 83–139.
matter neurons express less reelin and are abnormally Fernandez-Duque, D. and Posner, M.I. (2001) Brain imaging
distributed in schizophrenia: towards an integration of of attentional networks in normal and pathological states.
molecular and morphologic aspects of the neurodevelop- J. Clin. Exp. Neuropsychol., 23: 74–93.
mental hypothesis. Mol. Psychiatry, 8: 769, 821–731. Fish, B., Marcus, J., Hans, S.L., Auerbach, J.G. and Perdue, S.
Eastwood, S.L., Law, A.J., Everall, I.P. and Harrison, P.J. (1992) Infants at risk for schizophrenia: sequelae of a genetic
(2003) The axonal chemorepellant semaphorin 3A is neurointegrative defect. A review and replication analysis of
increased in the cerebellum in schizophrenia and may pandysmaturation in the Jerusalem infant development
contribute to its synaptic pathology. Mol. Psychiatry, 8: Study. Arch. Gen. Psychiatry, 49: 221–235.
148–155. Frame, S. and Cohen, P. (2001) GSK3 takes centre
Eaton, W.W., Thara, R., Federman, B., Melton, B. and stage more than 20 years after its discovery. Biochem. J.,
Liang, K.Y. (1995) Structure and course of positive and 359: 1–16.
negative symptoms in schizophrenia. Arch. Gen. Psychiatry, Freedman, R., Waldo, M., Bickford-Wimer, P. and
52: 127–134. Nagamoto, H. (1991) Elementary neuronal dysfunctions in
Eisch, A.J. (2002) Adult neurogenesis: implications for schizophrenia. Schizophr. Res., 4: 233–243.
psychiatry. Prog. Brain Res., 138: 315–342. Friston, K.J. (1999) Schizophrenia and the disconnection
Ellenbroek, B.A. and Cools, A.R. (2000) Animal models for hypothesis. Acta Psychiatr. Scand. Suppl., 395: 68–79.
the negative symptoms of schizophrenia. Behav. Pharmacol., Frith, C.D. and Done, D.J. (1988) Towards a neuropsychology
11: 223–233. of schizophrenia. British Journal of Psychiatry, 153: 437–443.
Emamian, E.S., Hall, D., Birnbaum, M.J., Karayiorgou, M. Garey, L.J., Ong, W.Y., Patel, T.S., Kanani, M., Davis, A.,
and Gogos, J.A. (2004) Convergent evidence for impaired Mortimer, A.M., Barnes, T.R. and Hirsch, S.R. (1998)
AKT1-GSK3beta signaling in schizophrenia. Nat. Genet., Reduced dendritic spine density on cerebral cortical pyrami-
36: 131–137. dal neurons in schizophrenia [see comments]. J. Neurol.
Eriksson, P.S., Perfilieva, E., Bjork-Eriksson, T., Alborn, A.M., Neurosurg. Psychiatry, 65: 446–453.
Nordborg, C., Peterson, D.A. and Gage, F.H. (1998) Geddes, J.R. and Lawrie, S.M. (1995) Obstetric complications
Neurogenesis in the adult human hippocampus. Nat. Med., and schizophrenia: a meta-analysis. Br. J. Psychiatry, 167:
4: 1313–1317. 786–793.
Erritzoe, D., Talbot, P., Frankle, W.G. and Abi-Dargham, A. Glantz, L.A. and Lewis, D.A. (1995) Assessment of spine
(2003) Positron emission tomography and single photon density on layer III pyramidal cells in the prefrontal cortex of
emission CT molecular imaging in schizophrenia. schizophrenic subjects. Society for Neuroscience Abstracts,
Neuroimaging Clin. N. Am., 13: 817–832. 21: 239.
Erwin, R.J., Turetsky, B.I., Moberg, P., Gur, R.C. and Goldberg, T.E., David, A. and Gold, J.M. (2003)
Gur, R.E. (1998) P50 abnormalities in schizophrenia: Neurocognitive deficits in schizophrenia. In: Hirsch S.R.
relationship to clinical and neuropsychological indices of and Weinberger D.R. (Eds.), Schizophrenia. Blackwell
attention. Schizophr. Res., 33: 157–167. Publishing, Malden, MA, pp. 168–184.
338

Goldner, E.M., Hsu, L., Waraich, P. and Somers, J.M. (2002) Harrison, P.J. and Owen, M.J. (2003) Genes for schizophrenia?
Prevalence and incidence studies of schizophrenic disorders: a Recent findings and their pathophysiological implications.
systematic review of the literature. Can J. Psychiatry, 47: Lancet, 361: 417–419.
833–843. Hatten, M.E. (1999) Central nervous system neuronal migra-
Gourion, D., Goldberger, C., Bourdel, M.C., Jean Bayle, F., tion. Annu. Rev. Neurosci., 22: 511–539.
Loo, H. and Krebs, M.O. (2004) Minor physical anomalies in Hattori, E., Liu, C., Badner, J.A., Bonner, T.I., Christian, S.L.,
patients with schizophrenia and their parents: prevalence and Maheshwari, M., Detera-Wadleigh, S.D., Gibbs, R.A. and
pattern of craniofacial abnormalities. Psychiatry Res., 125: Gershon, E.S. (2003) Polymorphisms at the G72/G30 gene
21–28. locus, on 13q33, are associated with bipolar disorder in two
Grant, S.G. (2003) Synapse signalling complexes and net- independent pedigree series. Am. J. Hum. Genet., 72:
works: machines underlying cognition. Bioessays, 25: 1131–1140.
1229–1235. Haydon, P.G. (2001) GLIA: listening and talking to the
Gray, J.A., Feldon, J., Rawlins, J.N.P., Helmsley, D.R. and synapse. Nat. Rev. Neurosci., 2: 185–193.
Smith, A.D. (1991) The neuropsychology of schizophrenia. Hazlett, E.A., Buchsbaum, M.S., Kemether, E., Bloom, R.,
Behavioral Brain Sciences, 14: 1–84. Platholi, J., Brickman, A.M., Shihabuddin, L., Tang, C. and
Guidotti, A., Auta, J., Davis, J.M., Di-Giorgi-Gerevini, V., Byne, W. (2004) Abnormal glucose metabolism in the
Dwivedi, Y., Grayson, D.R., Impagnatiello, F., Pandey, G., mediodorsal nucleus of the thalamus in schizophrenia. Am.
Pesold, C., Sharma, R., Uzunov, D., Costa, E. and DiGiorgi J. Psychiatry, 161: 305–314.
Gerevini, V. (2000) Decrease in reelin and glutamic acid Hecker, E. (1871) Die hebephrenie. Arch. Pathol. Anat.
decarboxylase67 (GAD67) expression in schizophrenia and Physiol. Klin. Med., 52: 394–401.
bipolar disorder: a postmortem brain study. Arch. Gen. Hemby, S.E., Ginsberg, S.D., Brunk, B., Arnold, S.E.,
Psychiatry, 57: 1061–1069. Trojanowski, J.Q. and Eberwine, J.H. (2002) Gene expres-
Gur, R.C., Moelter, S.T. and Ragland, J.D. (2000) Learning sion profile for schizophrenia: discrete neuron transcrip-
and memory in schizophrenia. In: Sharma T. and Harvey P. tion patterns in the entorhinal cortex. Arch. Gen. Psychiatry,
(Eds.), Cogntition in Schizophrenia: Impairments, Impor- 59: 631–640.
tance, and Treatment Strategies. Oxford University Press, Hetherington, P.A., Austin, K.B. and Shapiro, M.L. (1994)
New York, pp. 73–91. Ipsilateral associational pathway in the dentate gyrus: an
Gur, R.E., McGrath, C., Chan, R.M., Schroeder, L., excitatory feedback system that supports N-methyl-D-
Turner, T., Turetsky, B.I., Kohler, C., Alsop, D., aspartate-dependent long-term potentiation. Hippocampus,
Maldjian, J., Ragland, J.D. and Gur, R.C. (2002) An fMRI 4: 422–438.
study of facial emotion processing in patients with schizo- Hirayasu, Y., Asato, N., Ohta, H., Hokama, H., Arakaki, H.
phrenia. Am. J. Psychiatry, 159: 1992–1999. and Ogura, C. (1998) Abnormalities of auditory event-related
Gur, R.E., Turetsky, B.I., Bilker, W.B. and Gur, R.C. (1999) potentials in schizophrenia prior to treatment. Biol.
Reduced gray matter volume in schizophrenia. Arch. Gen. Psychiatry, 43: 244–253.
Psychiatry, 56: 905–911. Hof, P.R., Haroutunian, V., Copland, C., Davis, K.L. and
Hafner, H. and Heiden, Wad. (2003) Course and outcome of Buxbaum, J.D. (2002) Molecular and cellular evidence for an
schizophrenia. In: Hirsch S.R. and Weinberger D.R. (Eds.), oligodendrocyte abnormality in schizophrenia. Neurochem.
Schizophrenia. Blackwell Publishing, Malden, MA, Res., 27: 1193–1200.
pp. 101–141. Hof, P.R., Haroutunian, V., Friedrich, V.L., Jr., Byne, W.,
Hakak, Y., Walker, J.R., Li, C., Wong, W.H., Davis, K.L., Buitron, C., Perl, D.P. and Davis, K.L. (2003) Loss and
Buxbaum, J.D., Haroutunian, V. and Fienberg, A.A. (2001) altered spatial distribution of oligodendrocytes in the
Genome-wide expression analysis reveals dysregulation of superior frontal gyrus in schizophrenia. Biol. Psychiatry,
myelination-related genes in chronic schizophrenia. Proc. 53: 1075–1085.
Natl. Acad. Sci. USA, 98: 4746–4751. Hofer, A., Weiss, E.M., Golaszewski, S.M., Siedentopf, C.M.,
Hall, A.C., Lucas, F.R. and Salinas, P.C. (2000) Brinkhoff, C., Kremser, C., Felber, S. and
Axonal remodeling and synaptic differentiation in the Fleischhacker, W.W. (2003) An FMRI study of episodic
cerebellum is regulated by WNT-7a signaling. Cell, 100: encoding and recognition of words in patients with schizo-
525–535. phrenia in remission. Am. J. Psychiatry, 160: 911–918.
Harrison, P.J. (1999) The neuropathology of schizophrenia. Honer, W.G., Young, C. and Falkai, P. (2000) Synaptic
A critical review of the data and their interpretation. Brain, pathology. In: Harrison P.J. and Roberts G.W. (Eds.), The
122: 593–624. Neuropathology of Schizophrenia. Oxford University Press,
Harrison, P.J. and Eastwood, S.L. (2001) Neuropathological New York, pp. 105–136.
studies of synaptic connectivity in the hippocampal forma- Huber, K.M., Kayser, M.S. and Bear, M.F. (2000) Role
tion in schizophrenia. Hippocampus, 11: 508–519. for rapid dendritic protein synthesis in hippocampal
339

mGluR-dependent long-term depression. Science, 288: Kang, H. and Schuman, E.M. (1996) A requirement for local
1254–1257. protein synthesis in neurotrophin-induced hippocampal
Hwu, H.G., Liu, C.M., Fann, C.S., Ou-Yang, W.C. and synaptic plasticity. Science, 273: 1402–1406.
Lee, S.F. (2003) Linkage of schizophrenia with chromosome Karoumi, B., Laurent, A., Rosenfeld, F., Rochet, T.,
1q loci in Taiwanese families. Mol. Psychiatry, 8: 445–452. Brunon, A.M., Dalery, J., d’Amato, T. and Saoud, M.
Impagnatiello, F., Pisu, M.G., Pesold, C., Uzunov, D.P., (2000) Alteration of event related potentials in siblings
Dwivedi, Y., Pandey, G., Caruncho, H., Costa, E. and discordant for schizophrenia. Schizophr. Res., 41: 325–334.
Guidotti, A. (1998) Reelin expression is decreased in several Kathmann, N., von Recum, S., Haag, C. and Engel, R.R.
areas of schizophrenic brain. Society for Neuroscience (2000) Electrophysiological evidence for reduced latent
Abstracts, 24: 1275. inhibition in schizophrenic patients. Schizophr. Res., 45:
Iwata, N., Suzuki, T., Ikeda, M., Kitajima, T., Yamanouchi, Y., 103–114.
Inada, T. and Ozaki, N. (2004) No association with the Kim, J.J., Kwon, J.S., Park, H.J., Youn, T., Kang do, H.,
neuregulin 1 haplotype to Japanese schizophrenia. Mol. Kim, M.S., Lee, D.S. and Lee, M.C. (2003) Functional
Psychiatry, 9: 126–127. disconnection between the prefrontal and parietal cortices
Jackson, M.B. and Scharfman, H.E. (1996) Positive feedback during working memory processing in schizophrenia:
from hilar mossy cells to granule cells in the dentate gyrus a[15(O)]H2O PET study. Am. J. Psychiatry, 160: 919–923.
revealed by voltage-sensitive dye and microelectrode record- Kim, J.S., Kornhuber, H.H., Schmid-Burgk, W. and
ing. J. Neurophysiol., 76: 601–616. Holzmuller, B. (1980) Low cerebrospinal fluid glutamate in
Jacquet, H., Raux, G., Thibaut, F., Hecketsweiler, B., Houy, E., schizophrenic patients and a new hypothesis on schizo-
Demilly, C., Haouzir, S., Allio, G., Fouldrin, G., Drouin, V., phrenia. Neuroscience Letters, 30: 379–382.
Bou, J., Petit, M., Campion, D. and Frebourg, T. (2002) Kirkpatrick, B., Conley, R.C., Kakoyannis, A., Reep, R.L. and
PRODH mutations and hyperprolinemia in a subset of Roberts, R.C. (1999) Interstitial cells of the white matter in
schizophrenic patients. Hum. Mol. Genet., 11: 2243–2249. the inferior parietal cortex in schizophrenia: an unbiased
Jakob, H. and Beckmann, H. (1986) Prenatal developmental cell-counting study. Synapse, 34: 95–102.
disturbances in the limbic allocortex in schizophrenics. Kolomeets, N.S. and Uranova, N.A. (1999) Synaptic contacts
Journal of Neural Transmission, 65: 303–326. in schizophrenia: studies using immunocytochemical identi-
Jakob, H. and Beckmann, H. (1989) Gross and histological fication of dopaminergic neurons. Neurosci. Behav. Physiol.,
criteria for deveopmental disorders in brains of schizo- 29: 217–221.
phrenics. Journal of the Royal Society of Medicine, 82: Konradi, C. and Heckers, S. (2003) Molecular aspects of
466–469. glutamate dysregulation: implications for schizophrenia and
Jakob, H. and Beckmann, H. (1994) Cicumscribed malforma- its treatment. Pharmacol. Ther., 97: 153–179.
tion and nerve cell alterations in the entorhinal cortex of Kovelman, J.A. and Scheibel, A.B. (1984) A neurohistological
schizophrenics. Pathogenetic and clinical aspects. Journal of correlate of schizophrenia. Biological Psychiatry, 19:
Neural Transmission, 98: 83–106. 1601–1621.
Jessen, F., Scheef, L., Germeshausen, L., Tawo, Y., Kozlovsky, N., Belmaker, R.H. and Agam, G. (2000) Low
Kockler, M., Kuhn, K.U., Maier, W., Schild, H.H. and GSK-3beta immunoreactivity in postmortem frontal cortex
Heun, R. (2003) Reduced hippocampal activation during of schizophrenic patients. Am. J. Psychiatry, 157: 831–833.
encoding and recognition of words in schizophrenia patients. Kozlovsky, N., Belmaker, R.H. and Agam, G. (2001) Low
Am. J. Psychiatry, 160: 1305–1312. GSK-3 activity in frontal cortex of schizophrenic patients.
Jones, P., Rodgers, B., Murray, R. and Marmot, M. (1994) Schizophr. Res., 52: 101–105.
Child development risk factors for adult onset schizophrenia Kozlovsky, N., Belmaker, R.H. and Agam, G. (2002) GSK-3
in the British 1946 birth cohort. Lancet, 344: 1398–1402. and the neurodevelopmental hypothesis of schizophrenia.
Jonsson, E.G., Flyckt, L., Burgert, E., Crocq, M.A., Eur. Neuropsychopharmacol., 12: 13–25.
Forslund, K., Mattila-Evenden, M., Rylander, G., Kraepelin, E. (1919) Dementia Praecox and Paraphrenia.
Asberg, M., Nimgaonkar, V.L., Edman, G., Livingstone, Edinburgh.
Bjerkenstedt, L., Wiesel, F.A. and Sedvall, G.C. (2003) Krimer, L.S., Herman, M.M., Saunders, R.C., Boyd, J.C.,
Dopamine D3 receptor gene Ser9Gly variant and schizo- Hyde, T.M., Carter, J.M., Kleinman, J.E. and
phrenia: association study and meta-analysis. Psychiatr. Weinberger, D.R. (1997) A qualitative and quantitative
Genet., 13: 1–12. analysis of the entorhinal cortex in schizophrenia. Cerebral
Joyal, C.C., Laakso, M.P., Tiihonen, J., Syvalahti, E., Cortex, 7: 732–739.
Vilkman, H., Laakso, A., Alakare, B., Rakkolainen, V., Kung, L., Conley, R., Chute, D.J., Smialek, J. and
Salokangas, R.K. and Hietala, J. (2002) A volumetric MRI Roberts, R.C. (1998) Synaptic changes in the striatum of
study of the entorhinal cortex in first episode neuroleptic- schizophrenic cases: a controlled postmortem ultrastructural
naive schizophrenia. Biol. Psychiatry, 51: 1005–1007. study. Synapse, 28: 125–139.
340

Lauer, M., Beckmann, H. and Senitz, D. (2003) Increased Liu, W.S., Pesold, C., Rodriguez, M.A., Carboni, G., Auta, J.,
frequency of dentate granule cells with basal dendrites in the Lacor, P., Larson, J., Condie, B.G., Guidotti, A. and
hippocampal formation of schizophrenics. Psychiatry Res., Costa, E. (2001) Down-regulation of dendritic spine and
122: 89–97. glutamic acid decarboxylase 67 expressions in the reelin
Lemke, G. (2001) Glial control of neuronal development. haploinsufficient heterozygous reeler mouse. Proc. Natl.
Annu. Rev. Neurosci., 24: 87–105. Acad Sci. USA, 98: 3477–3482.
Lewis, C.M., Levinson, D.F., Wise, L.H., DeLisi, L.E., Lohr, J.B. and Flynn, K. (1993) Minor physical anomalies in
Straub, R.E., Hovatta, I., Williams, N.M., Schwab, S.G., schizophrenia and mood disorders. Schizophr. Bull., 19:
Pulver, A.E., Faraone, S.V., Brzustowicz, L.M., 551–556.
Kaufmann, C.A., Garver, D.L., Gurling, H.M., MacDonald, A.W. 3rd. and Carter, C.S. (2003) Event-related
Lindholm, E., Coon, H., Moises, H.W., Byerley, W., FMRI study of context processing in dorsolateral prefrontal
Shaw, S.H., Mesen, A., Sherrington, R., O’Neill, F.A., cortex of patients with schizophrenia. J. Abnorm. Psychol.,
Walsh, D., Kendler, K.S., Ekelund, J., Paunio, T., 112: 689–697.
Lonnqvist, J., Peltonen, L., O’Donovan, M.C., Owen, M.J., Malmberg, A., Lewis, G., David, A. and Allebeck, P. (1998)
Wildenauer, D.B., Maier, W., Nestadt, G., Blouin, J.L., Premorbid adjustment and personality in people with
Antonarakis, S.E., Mowry, B.J., Silverman, J.M., schizophrenia. Br. J. Psychiatry, 172: 308–313; discussion
Crowe, R.R., Cloninger, C.R., Tsuang, M.T., 314–305.
Malaspina, D., Harkavy-Friedman, J.M., Svrakic, D.M., Marcelis, M., Navarro-Mateu, F., Murray, R., Selten, J.P. and
Bassett, A.S., Holcomb, J., Kalsi, G., McQuillin, A., Van Os, J. (1998) Urbanization and psychosis: a study of
Brynjolfson, J., Sigmundsson, T., Petursson, H., Jazin, E., 1942–1978 birth cohorts in The Netherlands. Psychol. Med.,
Zoega, T. and Helgason, T. (2003) Genome scan meta- 28: 871–879.
analysis of schizophrenia and bipolar disorder, part II: Marenco, S. and Weinberger, D.R. (2000) The neurodevelop-
schizophrenia. Am. J. Hum. Genet, 73: 34–48. mental hypothesis of schizophrenia: following a trail of
Lewis, D.A. and Glantz, L.A. (1997) Specificity of decreased evidence from cradle to grave. Dev. Psychopathol., 12:
spine density on layer III pyramidal cells in schizophrenia. 501–527.
Schizophrenia Research, 24: 39. Markus, A., Zhong, J. and Snider, W.D. (2002) Raf and akt
Li, B.S., Ma, W., Jaffe, H., Zheng, Y., Takahashi, S., Zhang, L., mediate distinct aspects of sensory axon growth. Neuron, 35:
Kulkarni, A.B. and Pant, H.C. (2003a) Cyclin-dependent 65–76.
kinase-5 is involved in neuregulin-dependent activation of Matthies, H., Schroder, H., Wagner, M., Hollt, V. and
phosphatidylinositol 3-kinase and Akt activity mediating Krug, M. (1995) NMDA/R1-antisense oligonucleotide
neuronal survival. J. Biol. Chem., 278: 35702–35709. influences the early stage of long-term potentiation in the
Li, W., Zhang, Q., Oiso, N., Novak, E.K., Gautam, R., CA1-region of rat hippocampus. Neurosci. Lett., 202:
O’Brien, E.P., Tinsley, C.L., Blake, D.J., Spritz, R.A., 113–116.
Copeland, N.G., Jenkins, N.A., Amato, D., Roe, B.A., McCarley, R.W., Wible, C.G., Frumin, M., Hirayasu, Y.,
Starcevic, M., Dell’Angelica, E.C., Elliott, R.W., Mishra, V., Levitt, J.J., Fischer, I.A. and Shenton, M.E. (1999) MRI
Kingsmore, S.F., Paylor, R.E. and Swank, R.T. (2003b) anatomy of schizophrenia. Biol. Psychiatry, 45: 1099–1119.
Hermansky-Pudlak syndrome type 7 (HPS-7) results from McClintock, B.W., Shannon-Weickert, C., Halim, N.D.,
mutant dysbindin, a member of the biogenesis of lysosome- Lipska, B.K., Herman, M.M., Weinberger, D.R.,
related organelles complex 1 (BLOC-1). Nat. Genet., 35: Kleinman, J.E. and Straub, R.E. (2003) Reduced expression
84–89. of dysbindin protein in the dorsolateral prefrontal cortex of
Liddle, P. and Pantelis, C. (2003) Brain imaging in schizo- patients with schizophrenia. Society for Neuroscience, 33:
phrenia. In: Hirsch S.R. and Weinberger D.R. (Eds.), 317–319.
Schizophrenia. Blackwell Publishing, Malden, MA, McNeely, H.E., West, R., Christensen, B.K. and Alain, C.
pp. 403–417. (2003) Neurophysiological evidence for disturbances of
Lin, C.H., Yeh, S.H., Lu, K.T., Leu, T.H., Chang, W.C. and conflict processing in patients with schizophrenia. J.
Gean, P.W. (2001) A role for the PI-3 kinase signaling Abnorm. Psychol., 112: 679–688.
pathway in fear conditioning and synaptic plasticity in the McNeil, T.F., Cantor-Graae, E., Nordstrom, L.G. and
amygdala. Neuron, 31: 841–851. Rosenlund, T. (1996) Are reduced head circumference at
Liu, H., Heath, S.C., Sobin, C., Roos, J.L., Galke, B.L., birth and increased obstetric complications associated only
Blundell, M.L., Lenane, M., Robertson, B., Wijsman, E.M., with schizophrenic psychosis? A comparison with schizo-
Rapoport, J.L., Gogos, J.A. and Karayiorgou, M. (2002) affective and unspecified functional psychoses. Schizophr.
Genetic variation at the 22q11 PRODH2/DGCR6 locus Res., 22: 41–47.
presents an unusual pattern and increases susceptibility to Mednick, S.A., Machon, R.A., Huttunen, M.O. and
schizophrenia. Proc. Natl. Acad. Sci. USA, 99: 3717–3722. Bonnett, D. (1988) Adult schizophrenia following prenatal
341

exposure to an influenza epidemic. Archives of General Moberg, P.J. and Turetsky, B.I. (2003) Scent of a disorder:
Psychiatry, 45: 189–192. olfactory functioning in schizophrenia. Curr. Psychiatry
Menon, V., Anagnoson, R.T., Glover, G.H. and Rep., 5: 311–319.
Pfefferbaum, A. (2001) Functional magnetic resonance Moises, H.W., Zoega, T. and Gottesman, I.I. (2002) The glial
imaging evidence for disrupted basal ganglia function in growth factors deficiency and synaptic destabilization
schizophrenia. Am. J. Psychiatry, 158: 646–649. hypothesis of schizophrenia. BMC Psychiatry, 2: 8.
Meyer-Lindenberg, A., Poline, J.B., Kohn, P.D., Holt, J.L., Morris, D.W., McGhee, K.A., Schwaiger, S., Scully, P.,
Egan, M.F., Weinberger, D.R. and Berman, K.F. (2001) Quinn, J., Meagher, D., Waddington, J.L., Gill, M. and
Evidence for abnormal cortical functional connectivity Corvin, A.P. (2003) No evidence for association of the
during working memory in schizophrenia. Am. J. dysbindin gene [DTNBP1] with schizophrenia in an Irish
Psychiatry, 158: 1809–1817. population-based study. Schizophr. Res., 60: 167–172.
Millar, J.K., Christie, S., Anderson, S., Lawson, D., Hsiao-Wei Morris, J.K., Lin, W., Hauser, C., Marchuk, Y., Getman, D.
Loh, D., Devon, R.S., Arveiler, B., Muir, W.J., and Lee, K.F. (1999) Rescue of the cardiac defect in
Blackwood, D.H. and Porteous, D.J. (2001) Genomic ErbB2 mutant mice reveals essential roles of ErbB2 in
structure and localisation within a linkage hotspot of peripheral nervous system development. Neuron, 23:
Disrupted In Schizophrenia 1, a gene disrupted by a 273–283.
translocation segregating with schizophrenia. Mol. Mortensen, P.B., Pedersen, C.B., Westergaard, T.,
Psychiatry, 6: 173–178. Wohlfahrt, J., Ewald, H., Mors, O., Andersen, P.K. and
Millar, J.K., Wilson-Annan, J.C., Anderson, S., Christie, S., Melbye, M. (1999) Effects of family history and place and
Taylor, M.S., Semple, C.A., Devon, R.S., Clair, D.M., season of birth on the risk of schizophrenia. N. Engl. J. Med.,
Muir, W.J., Blackwood, D.H. and Porteous, D.J. (2000) 340: 603–608.
Disruption of two novel genes by a translocation Murray, C.J.L., Lopez, A.D., Murray, C.J.L., Lopez, A.D.,
co-segregating with schizophrenia. Hum. Mol. Genet., 9: Murray, C.J.L. and Lopez, A.D.S. (1996) The Global Burden
1415–1423. of Disease. Harvard University Press, Boston.
Mirnics, K., Middleton, F.A., Marquez, A., Lewis, D.A. and Nakazawa, K., Quirk, M.C., Chitwood, R.A., Watanabe, M.,
Levitt, P. (2000) Molecular characterization of schizophrenia Yeckel, M.F., Sun, L.D., Kato, A., Carr, C.A., Johnston, D.,
viewed by microarray analysis of gene expression in Wilson, M.A. and Tonegawa, S. (2002) Requirement for
prefrontal cortex. Neuron, 28: 53–67. hippocampal CA3 NMDA receptors in associative memory
Mirnics, K., Middleton, F.A., Stanwood, G.D., Lewis, D.A. recall. Science, 297: 211–218.
and Levitt, P. (2001) Disease-specific changes in regulator of Niemi, L.T., Suvisaari, J.M., Tuulio-Henriksson, A. and
G-protein signaling 4 (RGS4) expression in schizophrenia. Lonnqvist, J.K. (2003) Childhood developmental abnormal-
Mol. Psychiatry, 6: 293–301. ities in schizophrenia: evidence from high-risk studies.
Miyakawa, T., Leiter, L.M., Gerber, D.J., Gainetdinov, R.R., Schizophr. Res., 60: 239–258.
Sotnikova, T.D., Zeng, H., Caron, M.G. and Tonegawa, S. Nowakowski, R.S. and Rakic, P. (1981) The site of origin and
(2003) Conditional calcineurin knockout mice exhibit multi- route and rate of migration of neurons to the hippocampal
ple abnormal behaviors related to schizophrenia. Proc. Natl. region in the rhesus monkey. Journal of Comparative
Acad. Sci. USA, 100: 8987–8992. Neurology, 196: 129–154.
Miyamoto, S., Stroup, T.S., Duncan, G.E., Aoba, A. and O’Callaghan, E., Larkin, C., Kinsella, A. and Waddington, J.L.
Lieberman, J.A. (2003) Acute pharmacological treatment of (1991) Familial, obstetric, and other clinical correlates of
schizophrenia. In: Hirsch S.R. and Weinberger D.R. (Eds.), minor physical anomalies in schizophrenia. American
Schizophrenia. Blackwell Publishing, Malden, MA, Journal of Psychiatry, 148: 479–483.
pp. 442–473. O’Donnell, B.F., McCarley, R.W., Potts, G.F., Salisbury, D.F.,
Miyaoka, T., Seno, H. and Ishino, H. (1999) Increased Nestor, P.G., Hirayasu, Y., Niznikiewicz, M.A., Barnard, J.,
expression of Wnt-1 in schizophrenic brains. Schizophr. Shen, Z.J., Weinstein, D.M., Bookstein, F.L. and
Res., 38: 1–6. Shenton, M.E. (1999) Identification of neural circuits under-
Mizuno, M., Yamada, K., Takei, N., Tran, M.H., He, J., lying P300 abnormalities in schizophrenia. Psychophysiol-
Nakajima, A., Nawa, H. and Nabeshima, T. (2003) ogy, 36: 388–398.
Phosphatidylinositol 3-kinase: a molecule mediating O’Donovan, M.C., Williams, N.M. and Owen, M.J. (2003)
BDNF-dependent spatial memory formation. Mol. Recent advances in the genetics of schizophrenia. Hum.
Psychiatry, 8: 217–224. Mol. Genet., 12 Spec No 2: R125–R133.
Moberg, P.J., Agrin, R., Gur, R.E., Gur, R.C., Turetsky, B.I. Ohnuma, T., Kimura, M., Takahashi, T., Iwamoto, N. and
and Doty, R.L. (1999) Olfactory dysfunction in schizophre- Arai, H. (1997) A magnetic resonance imaging study in
nia: a qualitative and quantitative review. Neuropsycho- first-episode disorganized-type patients with schizophrenia.
pharmacology, 21: 325–340. Psychiatry Clin. Neurosci., 51: 9–15.
342

Olin, S.C., Mednick, S.A., Cannon, T., Jacobsen, B., Parnas, J., Riethmacher, D., Sonnenberg-Riethmacher, E., Brinkmann, V.,
Schulsinger, F. and Schulsinger, H. (1998) School teacher Yamaai, T., Lewin, G.R. and Birchmeier, C. (1997) Severe
ratings predictive of psychiatric outcome 25 years later. Br. J. neuropathies in mice with targeted mutations in the ErbB3
Psychiatry Suppl., 172: 7–13. receptor. Nature, 389: 725–730.
Olney, J.W., Newcomer, J.W. and Farber, N.B. (1999) NMDA Rioux, L., Nissanov, J., Lauber, K., Bilker, W.B. and
receptor hypofunction model of schizophrenia. J. Psychiatr. Arnold, S.E. (2003) Distribution of microtubule-associated
Res., 33: 523–533. protein MAP2-immunoreactive interstitial neurons in the
Owen, M.J., O’Donovan, M.C. and Gottesman, I.I. (2002) parahippocampal white matter in subjects with schizophre-
Schizophrenia. In: McGuffin P., Owen M.J. and nia. Am. J. Psychiatry, 160: 149–155.
Gottesman I.I. (Eds.), Psychiatric Genetics and Genomics. Roberts, R.C., Conley, R., Kung, L., Peretti, F.J. and
Oxford University Press, New York, pp. 247–266. Chute, D.J. (1996) Reduced striatal spine size in schizo-
Ozaki, M., Sasner, M., Yano, R., Lu, H.S. and Buonanno, A. phrenia: a postmortem ultrastructural study. Neuroreport, 7:
(1997) Neuregulin-beta induces expression of an NMDA- 1214–1218.
receptor subunit. Nature, 390: 691–694. Rosoklija, G., Kaufman, M.A., Liu, D., Hays, A.P., Latov, N.,
Ozaki, M., Tohyama, K., Kishida, H., Buonanno, A., Yano, R. Waniek, C., Keilp, J.G., Wu, A., Sadiq, S.A., Gorman, J.,
and Hashikawa, T. (2000) Roles of neuregulin in synapto- Prohovnik, I. and Dwork, A.J. (1995) Subicular MAP-2
genesis between mossy fibers and cerebellar granule cells. immunoreactivity in schizophrenia. Society for Neuroscience
J. Neurosci. Res., 59: 612–623. Abstracts, 21: 2126.
Pappas, G.D., Kriho, V. and Pesold, C. (2001) Reelin in the Rosoklija, G., Toomayan, G., Ellis, S.P., Keilp, J., Mann, J.J.,
extracellular matrix and dendritic spines of the cortex and Latov, N., Hays, A.P. and Dwork, A.J. (2000) Structural
hippocampus: a comparison between wild type and hetero- abnormalities of subicular dendrites in subjects with
zygous reeler mice by immunoelectron microscopy. schizophrenia and mood disorders: preliminary findings.
J. Neurocytol., 30: 413–425. Arch. Gen. Psychiatry, 57: 349–356.
Paradiso, S., Andreasen, N.C., Crespo-Facorro, B., Saint-Cyr, J.A. (2003) Frontal-striatal circuit functions:
O’Leary, D.S., Watkins, G.L., Boles Ponto, L.L. and context, sequence, and consequence. J. Int. Neuropsychol.
Hichwa, R.D. (2003) Emotions in unmedicated patients Soc., 9: 103–127.
with schizophrenia during evaluation with positron emission Salinas, P.C. (2003) Synaptogenesis: Wnt and TGF-beta take
tomography. Am. J. Psychiatry, 160: 1775–1783. centre stage. Curr. Biol., 13: R60–R62.
Pekkonen, E., Katila, H., Ahveninen, J., Karhu, J., Sanna, P.P., Cammalleri, M., Berton, F., Simpson, C.,
Huotilainen, M. and Tiihonen, J. (2002) Impaired temporal Lutjens, R., Bloom, F.E. and Francesconi, W. (2002)
lobe processing of preattentive auditory discrimination in Phosphatidylinositol 3-kinase is required for the expression
schizophrenia. Schizophr. Bull., 28: 467–474. but not for the induction or the maintenance of long-term
Pesold, C., Impagnatiello, F., Pisu, M.G., Uzunov, D.P., potentiation in the hippocampal CA1 region. J. Neurosci.,
Costa, E., Guidotti, A. and Caruncho, H.J. (1998) Reelin 22: 3359–3365.
is preferentially expressed in neurons synthesizing gamma- Sarnat, H.B. and Flores-Sarnat, L. (2002) Role of Cajal-Retzius
aminobutyric acid in cortex and hippocampus of adult rats. and subplate neurons in cerebral cortical development.
Proc. Natl. Acad. Sci. USA, 95: 3221–3226. Semin. Pediatr. Neurol., 9: 302–308.
Pessoa, L., Kastner, S. and Ungerleider, L.G. (2003) Saykin, A.J., Gur, R.C., Gur, R.E., Mozley, D., Mozley, L.H.,
Neuroimaging studies of attention: from modulation of Resnick, S.M., Kester, B. and Stafiniak, P. (1991)
sensory processing to top-down control. J. Neurosci., 23: Neuropsychological function in schizophrenia: selective
3990–3998. impairment in memory and learning. Archives of General
Petrozzino, J.J. and Connor, J.A. (1994) Dendritic Ca2þ Psychiatry, 48: 618–624.
accumulations and metabotropic glutamate receptor activa- Schiffman, J., Ekstrom, M., LaBrie, J., Schulsinger, F.,
tion associated with an N-methyl-D-aspartate receptor- Sorensen, H. and Mednick, S. (2002) Minor physical
independent long-term potentiation in hippocampal CA1 anomalies and schizophrenia spectrum disorders: a prospec-
neurons. Hippocampus, 4: 546–558. tive investigation. Am. J. Psychiatry, 159: 238–243.
Pilz, D., Stoodley, N. and Golden, J.A. (2002) Neuronal Schmid, R.S., McGrath, B., Berechid, B.E., Boyles, B.,
migration, cerebral cortical development, and cerebral Marchionni, M., Sestan, N. and Anton, E.S. (2003)
cortical anomalies. J. Neuropathol. Exp. Neurol., 61: 1–11. Neuregulin 1-erbB2 signaling is required for the establish-
Potkin, S.G., Alva, G., Fleming, K., Anand, R., Keator, D., ment of radial glia and their transformation into astrocytes
Carreon, D., Doo, M., Jin, Y., Wu, J.C. and Fallon, J.H. in cerebral cortex. Proc. Natl. Acad. Sci. USA, 100:
(2002) A PET study of the pathophysiology of negative 4251–4256.
symptoms in schizophrenia. Positron emission tomography. Schwab, S.G., Knapp, M., Mondabon, S., Hallmayer, J.,
Am. J. Psychiatry, 159: 227–237. Borrmann-Hassenbach, M., Albus, M., Lerer, B.,
343

Rietschel, M., Trixler, M., Maier, W. and Wildenauer, D.B. Stefansson, H., Steinthorsdottir, V., Thorgeirsson, T.E.,
(2003) Support for association of schizophrenia with genetic Gulcher, J.R. and Stefansson, K. (2004) Neuregulin 1 and
variation in the 6p22.3 gene, dysbindin, in sib-pair families schizophrenia. Ann. Med., 36: 62–71.
with linkage and in an additional sample of triad families. Stefansson, H., Thorgeirsson, T.E., Gulcher, J.R. and
Am. J. Hum. Genet., 72: 185–190. Stefansson, K. (2003b) Neuregulin 1 in schizophrenia: out
Schwob, J.E. (2002) Neural regeneration and the peripheral of Iceland. Mol. Psychiatry, 8: 639–640.
olfactory system. Anat. Rec., 269: 33–49. Straub, R.E., Jiang, Y., MacLean, C.J., Ma, Y., Webb, B.T.,
Selemon, L.D., Rajkowska, G. and Goldman-Rakic, P.S. Myakishev, M.V., Harris-Kerr, C., Wormley, B., Sadek, H.,
(1998) Elevated neuronal density in prefrontal area 46 in Kadambi, B., Cesare, A.J., Gibberman, A., Wang, X.,
brains from schizophrenic patients: application of a three- O’Neill, F.A., Walsh, D. and Kendler, K.S. (2002) Genetic
dimensional, stereologic counting method. Journal of variation in the 6p22.3 gene DTNBP1, the human ortholog
Comparative Neurology, 392: 402–412. of the mouse dysbindin gene, is associated with schizophre-
Shah, N.M. and Anderson, D.J. (1997) Integration of multiple nia. Am. J. Hum. Genet., 71: 337–348.
instructive cues by neural crest stem cells reveals cell-intrinsic Susser, E., Neugebauer, R., Hoek, H.W., Brown, A.S., Lin, S.,
biases in relative growth factor responsiveness. Proc. Natl. Labovitz, D. and Gorman, J.M. (1996) Schizophrenia after
Acad. Sci. USA, 94: 11369–11374. prenatal famine. Archives of General Psychiatry, 53: 25–31.
Shah, N.M., Marchionni, M.A., Isaacs, I., Stroobant, P. and Talbot, K. and Arnold, S.E. (2002) The parahippocampal
Anderson, D.J. (1994) Glial growth factor restricts mamma- region in schizophrenia. In: Witter, M. and Wouterlood, F.
lian neural crest stem cells to a glial fate. Cell, 77: 349–360. (Eds.), The Parahippocampal Region, Organization and Role
Sheng, M. and Kim, E. (2000) The Shank family of scaffold in Cognitive Functions. Oxford University Press, Oxford,
proteins. J. Cell Sci., 113(Pt 11): 1851–1856. pp. 295–318.
Shi, Y.Y., Zhao, X.Z., Tang, J.X., Gu, N.F., Feng, G.Y., Talbot, K., Eidem, W.L., Tinsley, C.L., Benson, M.A.,
Zhu, S.M., Ji, L.P. and He, L. (2003) Non-family based Thompson, E.W., Smith, R.J., Hahn C-G, Siegel, S.J.,
association study of DTNBP1 in 6p22.3 and schizophrenia in Trojanowski, J.Q., Gur, R.E., Blake, D.J. and Arnold, S.E.
geographically and genetically structured Chinese Han (2004) Dysbindin is reduced in intrinsic, glutamatergic
population. In: Barden N., Côté M, DeLisi L., Gill M., terminals of the hippocampal formation in schizophrenia.
Kelsoe J. and Schalling M. (Eds.), XIth World Congress Journal of Clinical Investigation, (in press).
of Psychiatric Genetics, Quebec, 102 p. Tang, J.X., Chen, W.Y., He, G., Zhou, J., Gu, N.F., Feng, G.Y.
Sivkov, S.T. and Akabaliev, V.H. (2003) Minor physical and He, L. (2004) Polymorphisms within 50 end of the
anomalies in mentally healthy subjects: internal consistency Neuregulin 1 gene are genetically associated with
of the Waldrop physical anomaly scale. Am. J. Human Biol., schizophrenia in the Chinese population. Mol. Psychiatry,
15: 61–67. 9: 11–12.
Soustek, Z. (1989) Ultrastructure of cortical synapses in the Tang, J.X., Zhou, J., Fan, J.B., Li, X.W., Shi, Y.Y., Gu, N.F.,
brain of schizophrenics. Zentralblatt allegmenaine Pathologie Feng, G.Y., Xing, Y.L., Shi, J.G. and He, L. (2003) Family-
und pathologische Anatomie, 135: 25–32. based association study of DTNBP1 in 6p22.3 and schizo-
Stefansson, H., Sarginson, J., Kong, A., Yates, P., phrenia. Molecular Psychiatry, 8: 717–718.
Steinthorsdottir, V., Gudfinnsson, E., Gunnarsdottir, S., Tissir, F. and Goffinet, A.M. (2003) Reelin and brain
Walker, N., Petursson, H., Crombie, C., Ingason, A., development. Nat. Rev. Neurosci., 4: 496–505.
Gulcher, J.R., Stefansson, K. and St. Clair, D. (2003a) Torrey, E.F., Miller, J., Rawlings, R. and Yolken, R.H. (1997)
Association of neuregulin 1 with schizophrenia confirmed in Seasonality of births in schizophrenia and bipolar disorder:
a Scottish population. Am. J. Hum. Genet., 72: 83–87. a review of the literature. Schizophr. Res., 28: 1–38.
Stefansson, H., Sigurdsson, E., Steinthorsdottir, V., Turetsky, B., Colbath, E.A. and Gur, R.E. (1998a) P300
Bjornsdottir, S., Sigmundsson, T., Ghosh, S., subcomponent abnormalities in schizophrenia: II.
Brynjolfsson, J., Gunnarsdottir, S., Ivarsson, O., Chou, T.T., Longitudinal stability and relationship to symptom change.
Hjaltason, O., Birgisdottir, B., Jonsson, H., Biol. Psychiatry, 43: 31–39.
Gudnadottir, V.G., Gudmundsdottir, E., Bjornsson, A., Turetsky, B.I., Cannon, T.D. and Gur, R.E. (2000a) P300
Ingvarsson, B., Ingason, A., Sigfusson, S., Hardardottir, H., subcomponent abnormalities in schizophrenia: III. Deficits
Harvey, R.P., Lai, D., Zhou, M., Brunner, D., Mutel, V., in unaffected siblings of schizophrenic probands. Biol.
Gonzalo, A., Lemke, G., Sainz, J., Johannesson, G., Psychiatry, 47: 380–390.
Andresson, T., Gudbjartsson, D., Manolescu, A., Turetsky, B.I., Colbath, E.A. and Gur, R.E. (1998b)
Frigge, M.L., Gurney, M.E., Kong, A., Gulcher, J.R., P300 subcomponent abnormalities in schizophrenia: I.
Petursson, H. and Stefansson, K. (2002) Neuregulin 1 and Physiological evidence for gender and subtype
susceptibility to schizophrenia. Am. J. Hum. Genet., 71: specific differences in regional pathology. Biol. Psychiatry,
877–892. 43: 84–96.
344

Turetsky, B.I., Moberg, P.J., Owzar, K., Johnson, S.C., Weinberger D.R. (Eds.), Schizophrenia. Blackwell
Doty, R.L. and Gur, R.E. (2003) Physiologic impairment Publishing, Malden, MA, pp. 326–348.
of olfactory stimulus processing in schizophrenia. Biol. Weiss, E.M., Golaszewski, S., Mottaghy, F.M., Hofer, A.,
Psychiatry, 53: 403–411. Hausmann, A., Kemmler, G., Kremser, C., Brinkhoff, C.,
Turetsky, B.I., Moberg, P.J., Yousem, D.M., Doty, R.L., Felber, S.R. and Wolfgang Fleischhacker, W. (2003) Brain
Arnold, S.E. and Gur, R.E. (2000b) Reduced olfactory bulb activation patterns during a selective attention test-a func-
volume in patients with schizophrenia. Am. J. Psychiatry, tional MRI study in healthy volunteers and patients with
157: 828–830. schizophrenia. Psychiatry Res., 123: 1–15.
Uranova, N.A., Casanova, M.F., DeVaughn, N.M., Wible, C.G., Kubicki, M., Yoo, S.S., Kacher, D.F.,
Orlovskaya, D.D. and Denisov, D.V. (1996) Ultrastructural Salisbury, D.F., Anderson, M.C., Shenton, M.E.,
alterations of synaptic contacts and astrocytes in postmortem Hirayasu, Y., Kikinis, R., Jolesz, F.A. and McCarley, R.W.
caudate nucleus of schizophrenic patients. Schizophr. Res., (2001) A functional magnetic resonance imaging study of
22: 81–83. auditory mismatch in schizophrenia. Am. J. Psychiatry, 158:
Van Den Bogaert, A., Schumacher, J., Schulze, T.G., 938–943.
Otte, A.C., Ohlraun, S., Kovalenko, S., Becker, T., Williams, H.J., Williams, N., Spurlock, G., Norton, N.,
Freudenberg, J., Jonsson, E.G., Mattila-Evenden, M., Zammit, S., Kirov, G., Owen, M.J. and O’Donovan, M.C.
Sedvall, G.C., Czerski, P.M., Kapelski, P., Hauser, J., (2003a) Detailed analysis of PRODH and PsPRODH reveals
Maier, W., Rietschel, M., Propping, P., Nothen, M.M. and no association with schizophrenia. Am. J. Med. Genet.,
Cichon, S. (2003) The DTNBP1 (Dysbindin) gene contributes 120B: 42–46.
to schizophrenia, depending on family history of the disease. Williams, L.M., Gordon, E., Wright, J. and Bahramali, H.
Am. J. Hum. Genet., 73: 1438–1443. (2000) Late component ERPs are associated with
van den Oord, E.J., Sullivan, P.F., Jiang, Y., Walsh, D., three syndromes in schizophrenia. Int. J. Neurosci., 105:
O’Neill, F.A., Kendler, K.S. and Riley, B.P. (2003) 37–52.
Identification of a high-risk haplotype for the dystrobrevin Williams, N.M., Preece, A., Spurlock, G., Norton, N.,
binding protein 1 (DTNBP1) gene in the Irish study of Williams, H.J., Zammit, S., O’Donovan, M.C. and
high-density schizophrenia families. Mol. Psychiatry, 8: Owen, M.J. (2003b) Support for genetic variation in
499–510. neuregulin 1 and susceptibility to schizophrenia. Mol.
van Veen, V. and Carter, C.S. (2002) The anterior cingulate Psychiatry, 8: 485–487.
as a conflict monitor: fMRI and ERP studies. Physiol. Williams, N.M., Williams, H., Norton, N., Spurlock, G.,
Behav., 77: 477–482. Kirov, G., Morris, D.W., Waddington, J.L., Gill, M.,
Vaskovsky, A., Lupowitz, Z., Erlich, S. and Pinkas- Corvin, A.P., Owen, M.J. and O’Donovan, M.C. (2003c)
Kramarski, R. (2000) ErbB-4 activation promotes neurite Replication of schizophrenia susceptibility loci. In: Barden N.,
outgrowth in PC12 cells. J. Neurochem., 74: 979–987. Cote M., DeLisi L., Gill M., Kelsoe J. and Schalling M.
Vawter, M.P., Barrett, T., Cheadle, C., Sokolov, B.P., (Eds.), XIth World Congress of Psychiatric Genetics,
Wood, W.H., Donovan, D.M., Webster, M., Freed, W.J. Quebec, pp. 16–17.
and Becker, K.G. (2001) Application of cDNA microarrays Witter, M.P., Wouterlood, F.G., Naber, P.A. and Van
to examine gene expression differences in schizophrenia. Haeften, T. (2000) Anatomical organization of the para-
Brain Res. Bull, 55: 641–650. hippocampal-hippocampal network. Ann. NY Acad. Sci.,
Waldrop, M.F., Pedersen, F.A. and Bell, R.Q. (1968) Minor 911: 1–24.
physical anomalies and behavior in preschool children. Child Woldeyesus, M.T., Britsch, S., Riethmacher, D., Xu, L.,
Dev., 39: 391–400. Sonnenberg-Riethmacher, E., Abou-Rebyeh, F., Harvey, R.,
Walker, E. (1994) Developmentally moderated expressions of Caroni, P. and Birchmeier, C. (1999) Peripheral nervous
the neuropathology underlying schizophrenia. Schizophrenia system defects in erbB2 mutants following genetic rescue
Bulletin, 20: 453–480. of heart development. Genes Dev., 13: 2538–2548.
Wall, P.M. and Messier, C. (2001) The hippocampal formation Yamada, K., Detera-Wadleigh, S.D., Iwayama-Shigeno, Y.,
— orbitomedial prefrontal cortex circuit in the atten- Toyota, T., Corona, W., Hattori, E., McMahon, F. and
tional control of active memory. Behav. Brain. Res., 127: Yoshikawa, T. (2003) Suggestive evidence of associa-
99–117. tion between the DTNBP1 gene and schizophrenia in
Wang, Q., Liu, L., Pei, L., Ju, W., Ahmadian, G., Lu, J., the Japanese samples. In: Barden N., Côté M., DeLisi L.,
Wang, Y., Liu, F. and Wang, Y.T. (2003) Control of Gill M., Kelsoe J. and Schalling M. (Eds.),
synaptic strength, a novel function of Akt. Neuron, 38: XIth World Congress of Psychiatric Genetics, Quebec, 102 p.
915–928. Yang, J.Z., Si, T.M., Ruan, Y., Ling, Y.S., Han, Y.H.,
Weinberger, D.R. and Marenco, S. (2003) Schizophrenia as a Wang, X.L., Zhou, M., Zhang, H.Y., Kong, Q.M., Liu, C.,
neurodevelopmental disorder. In: Hirsch S.R. and Zhang, D.R., Yu, Y.Q., Liu, S.Z., Ju, G.Z., Shu, L., Ma, D.L.
345

and Zhang, D. (2003) Association study of neuregulin 1 gene alterations in schizophrenia. American Journal of
with schizophrenia. Mol. Psychiatry, 8: 706–709. Psychiatry, 154: 812–818.
Zaidel, D.W., Esiri, M.M. and Harrison, P.J. (1997) Size, Zhao, J.J. and Lemke, G. (1998) Selective disruption of
shape, and orientation of neurons in the left and right neuregulin-1 function in vertebrate embryos using
hippocampus: investigations of normal asymmetries and ribozyme-tRNA transgenes. Development, 125: 1899–1907.

View publication stats

Potrebbero piacerti anche