Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell ; 165(5): 1238-1254, 2016 May 19.
Article in English | MEDLINE | ID: mdl-27118425

ABSTRACT

Cerebral organoids, three-dimensional cultures that model organogenesis, provide a new platform to investigate human brain development. High cost, variability, and tissue heterogeneity limit their broad applications. Here, we developed a miniaturized spinning bioreactor (SpinΩ) to generate forebrain-specific organoids from human iPSCs. These organoids recapitulate key features of human cortical development, including progenitor zone organization, neurogenesis, gene expression, and, notably, a distinct human-specific outer radial glia cell layer. We also developed protocols for midbrain and hypothalamic organoids. Finally, we employed the forebrain organoid platform to model Zika virus (ZIKV) exposure. Quantitative analyses revealed preferential, productive infection of neural progenitors with either African or Asian ZIKV strains. ZIKV infection leads to increased cell death and reduced proliferation, resulting in decreased neuronal cell-layer volume resembling microcephaly. Together, our brain-region-specific organoids and SpinΩ provide an accessible and versatile platform for modeling human brain development and disease and for compound testing, including potential ZIKV antiviral drugs.


Subject(s)
Brain/cytology , Cell Culture Techniques , Models, Biological , Organoids , Zika Virus/physiology , Bioreactors , Cell Culture Techniques/economics , Embryo, Mammalian , Embryonic Development , Humans , Induced Pluripotent Stem Cells , Neurogenesis , Neurons/cytology , Organoids/virology , Zika Virus Infection/physiopathology , Zika Virus Infection/virology
2.
Expert Rev Mol Med ; 25: e3, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36517884

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterised by gradual memory loss and declining cognitive and executive functions. AD is the most common cause of dementia, affecting more than 50 million people worldwide, and is a major health concern in society. Despite decades of research, the cause of AD is not well understood and there is no effective curative treatment so far. Therefore, there is an urgent need to increase understanding of AD pathophysiology in the hope of developing a much-needed cure. Dissecting the cellular and molecular mechanisms of AD pathogenesis has been challenging as the most commonly used model systems such as transgenic animals and two-dimensional neuronal culture do not fully recapitulate the pathological hallmarks of AD. The recent advent of three-dimensional human brain organoids confers unique opportunities to study AD in a humanised model system by encapsulating many aspects of AD pathology. In the present review, we summarise the studies of AD using human brain organoids that recapitulate the major pathological components of AD including amyloid-ß and tau aggregation, neuroinflammation, mitochondrial dysfunction, oxidative stress and synaptic and circuitry dysregulation. Additionally, the current challenges and future directions of the brain organoids modelling system are discussed.


Subject(s)
Alzheimer Disease , Animals , Humans , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/pathology , Neurons/metabolism , Neurons/pathology , Organoids/metabolism , Organoids/pathology
3.
PLoS Biol ; 15(7): e2001246, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28683067

ABSTRACT

Clinical and preclinical studies indicate that early postnatal exposure to anesthetics can lead to lasting deficits in learning and other cognitive processes. The mechanism underlying this phenomenon has not been clarified and there is no treatment currently available. Recent evidence suggests that anesthetics might cause persistent deficits in cognitive function by disrupting key events in brain development. The hippocampus, a brain region that is critical for learning and memory, contains a large number of neurons that develop in the early postnatal period, which are thus vulnerable to perturbation by anesthetic exposure. Using an in vivo mouse model we demonstrate abnormal development of dendrite arbors and dendritic spines in newly generated dentate gyrus granule cell neurons of the hippocampus after a clinically relevant isoflurane anesthesia exposure conducted at an early postnatal age. Furthermore, we find that isoflurane causes a sustained increase in activity in the mechanistic target of rapamycin pathway, and that inhibition of this pathway with rapamycin not only reverses the observed changes in neuronal development, but also substantially improves performance on behavioral tasks of spatial learning and memory that are impaired by isoflurane exposure. We conclude that isoflurane disrupts the development of hippocampal neurons generated in the early postnatal period by activating a well-defined neurodevelopmental disease pathway and that this phenotype can be reversed by pharmacologic inhibition.


Subject(s)
Anesthetics, Inhalation/toxicity , Cognitive Dysfunction/chemically induced , Hippocampus/drug effects , Isoflurane/toxicity , TOR Serine-Threonine Kinases/metabolism , Animals , Dendritic Spines/drug effects , Dendritic Spines/pathology , Environmental Exposure , Hippocampus/growth & development , Hippocampus/pathology , Mice , Neurons/drug effects , Neurons/pathology
4.
Dev Neurosci ; 37(4-5): 428-39, 2015.
Article in English | MEDLINE | ID: mdl-26087836

ABSTRACT

Neonatal hypoxic-ischemic injury (HI) results in widespread cerebral encephalopathy and affects structures that are essential for neurocognitive function, such as the hippocampus. The dentate gyrus contains a reservoir of neural stem and progenitor cells (NSPCs) that are critical for postnatal development and normal adult function of the hippocampus, and may also facilitate the recovery of function after injury. Using a neonatal mouse model of mild-to-moderate HI and immunohistochemical analysis of NSPC development markers, we asked whether these cells are vulnerable to HI and how they respond to both injury and hypothermic therapy. We found that cleaved caspase-3 labeling in the subgranular zone, where NSPCs are located, is increased by more than 30-fold after HI. The population of cells positive for both proliferating cell nuclear antigen and nestin (PCNA+Nes+), which represent primarily actively proliferating NSPCs, are acutely decreased by 68% after HI. The NSPC population expressing NeuroD1, a marker for NSPCs transitioning to become fate-committed neural progenitors, was decreased by 47%. One week after HI, there was a decrease in neuroblasts and immature neurons in the dentate gyrus, as measured by doublecortin (DCX) immunolabeling, and at the same time PCNA+Nes+ cell density was increased by 71%. NSPCs expressing Tbr2, which identifies a highly proliferative intermediate neural progenitor population, increased by 107%. Hypothermia treatment after HI partially rescues both the acute decrease in PCNA+Nes+ cell density at 1 day after injury and the chronic loss of DCX immunoreactivity and reduction in NeuroD1 cell density measured at 1 week after injury. Thus, we conclude that HI causes an acute loss of dentate gyrus NSPCs, and that hypothermia partially protects NSPCs from HI.


Subject(s)
Dentate Gyrus/pathology , Hypothermia, Induced , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/therapy , Neural Stem Cells/physiology , Neurogenesis/physiology , Animals , Animals, Newborn , Dentate Gyrus/cytology , Disease Models, Animal , Doublecortin Protein , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology
5.
J Neurophysiol ; 109(4): 1174-81, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23197456

ABSTRACT

Brief strong depolarization of cerebellar Purkinje cells produces a slow inward cation current [depolarization-induced slow current (DISC)]. Previous work has shown that DISC is triggered by voltage-sensitive Ca influx in the Purkinje cell and is attenuated by blockers of vesicular loading and fusion. Here, we have sought to characterize the ion channel(s) underlying the DISC conductance. While the brief depolarizing steps that triggered DISC were associated with a large Ca transient, the onset of DISC current corresponded only with the Ca transient decay phase. Furthermore, substitution of external Na with the impermeant cation N-methyl-d-glucamine produced a complete and reversible block of DISC, suggesting that the DISC conductance was not Ca permeant. Transient receptor potential cation channel, subfamily M, members 4 (TRPM4) and 5 (TRPM5) are nonselective cation channels that are opened by Ca transients but do not flux Ca. They are expressed in Purkinje cells of the posterior cerebellum, where DISC is large, and, in these cells, DISC is strongly attenuated by nonselective blockers of TRPM4/5. However, measurement of DISC currents in Purkinje cells derived from TRPM4 null, TRPM5 null, and double null mice as well as wild-type mice with TRPM4 short hairpin RNA knockdown showed a partial attenuation with 35-46% of current remaining. Thus, while the DISC conductance is Ca triggered, Na permeant, and Ca impermeant, suggesting a role for TRPM4 and TRPM5, these ion channels are not absolutely required for DISC.


Subject(s)
Action Potentials/physiology , Purkinje Cells/physiology , Action Potentials/drug effects , Animals , Calcium/metabolism , Meglumine/analogs & derivatives , Meglumine/pharmacology , Mice , Mice, Inbred C57BL , Purkinje Cells/metabolism , RNA, Small Interfering , Sodium/metabolism , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
6.
Curr Opin Neurobiol ; 18(1): 108-15, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18514504

ABSTRACT

New neurons are continuously generated in the dentate gyrus of the mammalian hippocampus and in the subventricular zone of the lateral ventricles throughout life. The origin of these new neurons is believed to be from multipotent adult neural stem cells. Aided by new methodologies, significant progress has been made in the characterization of neural stem cells and their development in the adult brain. Recent studies have also begun to reveal essential extrinsic and intrinsic molecular mechanisms that govern sequential steps of adult neurogenesis in the hippocampus and subventricular zone/olfactory bulb, from proliferation and fate specification of neural progenitors to maturation, navigation, and synaptic integration of the neuronal progeny. Future identification of molecular mechanisms and physiological functions of adult neurogenesis will provide further insight into the plasticity and regenerative capacity of the mature central nervous system.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Neurons/metabolism , Stem Cells/metabolism , Telencephalon/cytology , Telencephalon/metabolism , Animals , Cell Movement/genetics , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Humans , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Neurons/cytology , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Stem Cells/cytology
7.
Cell Rep ; 28(6): 1419-1428.e3, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31390557

ABSTRACT

Excitation-inhibition (E-I) imbalance is considered a hallmark of various neurodevelopmental disorders, including schizophrenia and autism. How genetic risk factors disrupt coordinated glutamatergic and GABAergic synapse formation to cause an E-I imbalance is not well understood. Here, we show that knockdown of Disrupted-in-schizophrenia 1 (DISC1), a risk gene for major mental disorders, leads to E-I imbalance in mature dentate granule neurons. We found that excessive GABAergic inputs from parvalbumin-, but not somatostatin-, expressing interneurons enhance the formation of both glutamatergic and GABAergic synapses in immature mutant neurons. Following the switch in GABAergic signaling polarity from depolarizing to hyperpolarizing during neuronal maturation, heightened inhibition from excessive parvalbumin+ GABAergic inputs causes loss of excitatory glutamatergic synapses in mature mutant neurons, resulting in an E-I imbalance. Our findings provide insights into the developmental role of depolarizing GABA in establishing E-I balance and how it can be influenced by genetic risk factors for mental disorders.


Subject(s)
Genetic Predisposition to Disease , Mental Disorders/genetics , Neurons/physiology , Synapses/physiology , gamma-Aminobutyric Acid/physiology , Animals , Cell Polarity , Female , GABAergic Neurons/physiology , Gene Knockdown Techniques , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/physiology , Neural Inhibition , Neurogenesis/genetics , Neurogenesis/physiology , Risk Factors , Synapses/genetics , Synaptic Potentials
8.
Commun Integr Biol ; 11(2): 1-4, 2018.
Article in English | MEDLINE | ID: mdl-30083288

ABSTRACT

Experiments conducted in non-human primates have recently provided new evidence supporting a longstanding concern that exposure to general anesthesia during late intrauterine life or early childhood can cause lasting cognitive deficits through harmful effects on brain development. The mammalian target of rapamycin (mTOR) signaling system plays a key role in both normal brain development and in a wide range of developmental disorders that are characterized by cognitive deficits. Intriguingly, our recently published work shows that anesthetics can chronically alter mTOR signaling in the hippocampal dentate gyrus and that normalization of mTOR signaling can prevent anesthesia-induced perturbation of structure and function. In this addendum, we briefly discuss the putative role of mTOR in developmental anesthetic neurotoxicity.

11.
Neuron ; 96(5): 1041-1054.e5, 2017 Dec 06.
Article in English | MEDLINE | ID: mdl-29103808

ABSTRACT

Mutations of DISC1 (disrupted-in-schizophrenia 1) have been associated with major psychiatric disorders. Despite the hundreds of DISC1-binding proteins reported, almost nothing is known about how DISC1 interacts with other proteins structurally to impact human brain development. Here we solved the high-resolution structure of DISC1 C-terminal tail in complex with its binding domain of Ndel1. Mechanistically, DISC1 regulates Ndel1's kinetochore attachment, but not its centrosome localization, during mitosis. Functionally, disrupting DISC1/Ndel1 complex formation prolongs mitotic length and interferes with cell-cycle progression in human cells, and it causes cell-cycle deficits of radial glial cells in the embryonic mouse cortex and human forebrain organoids. We also observed similar deficits in organoids derived from schizophrenia patient induced pluripotent stem cells (iPSCs) with a DISC1 mutation that disrupts its interaction with Ndel1. Our study uncovers a new mechanism of action for DISC1 based on its structure, and it has implications for how genetic insults may contribute to psychiatric disorders.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/physiology , Mitosis/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neurogenesis/genetics , Neurons/physiology , Animals , Carrier Proteins/chemistry , Cell Cycle , Female , HeLa Cells , Humans , Immunohistochemistry , Male , Mice , Models, Molecular , Nerve Tissue Proteins/chemistry , Neural Stem Cells , Neurons/pathology , Pluripotent Stem Cells , Pregnancy , Protein Binding , Schizophrenia/pathology
12.
Neurotoxicol Teratol ; 60: 33-39, 2017.
Article in English | MEDLINE | ID: mdl-27751818

ABSTRACT

The mechanism by which anesthetics might act on the developing brain in order to cause long term deficits remains incompletely understood. The hippocampus has been identified as a structure that is likely to be involved, as rodent models show numerous deficits in behavioral tasks of learning that are hippocampal-dependent. The hippocampus is an unusual structure in that it is the site of large amounts of neurogenesis postnatally, particularly in the first year of life in humans, and these newly generated neurons are critical to the function of this structure. Intriguingly, neurogenesis is a major developmental event that occurs during postulated windows of vulnerability to developmental anesthetic neurotoxicity across the different species in which it has been studied. In this review, we examine the evidence for anesthetic effects on neurogenesis in the early postnatal period and ask whether neurogenesis should be studied further as a putative mechanism of injury. Multiple anesthetics are considered, and both in vivo and in vitro work is presented. While there is abundant evidence that anesthetics act to suppress neurogenesis at several different phases, evidence of a causal link between these effects and any change in learning behavior remains elusive.


Subject(s)
Anesthetics/adverse effects , Brain/growth & development , Neurogenesis/drug effects , Neurotoxicity Syndromes/etiology , Animals , Humans
13.
Article in English | MEDLINE | ID: mdl-26801682

ABSTRACT

Psychiatric disorders continue to be among the most challenging disorders to diagnose and treat because there is no single genetic or anatomical locus that is causative for the disease. Current treatments are often blunt tools used to ameliorate the most severe symptoms, at the risk of disrupting functional neural systems. There is a critical need to develop new therapeutic strategies that can target circumscribed functional or anatomical domains of pathology. Adult hippocampal neurogenesis may be one such domain. Here, we review the evidence suggesting that adult hippocampal neurogenesis plays a role in emotional regulation and forms of learning and memory that include temporal and spatial memory encoding and context discrimination, and that its dysregulation is associated with psychiatric disorders, such as affective disorders, schizophrenia, and drug addiction. Further, adult neurogenesis has proven to be an effective model to investigate basic processes of neuronal development and converging evidence suggests that aberrant neural development may be an etiological factor, even in late-onset diseases. Constitutive neurogenesis in the hippocampus of the mature brain reflects large-scale plasticity unique to this region and could be a potential hub for modulation of a subset of cognitive and affective behaviors that are affected by multiple psychiatric disorders.


Subject(s)
Mental Disorders/pathology , Neurogenesis , Adult , Animals , Disease Models, Animal , Humans , Mental Disorders/psychology
14.
Nat Commun ; 72016 06 01.
Article in English | MEDLINE | ID: mdl-27249678

ABSTRACT

Several genome- and proteome-wide studies have associated transcription and translation changes of CRMP2 (collapsing response mediator protein 2) with psychiatric disorders, yet little is known about its function in the developing or adult mammalian brain in vivo. Here we show that brain-specific Crmp2 knockout (cKO) mice display molecular, cellular, structural and behavioural deficits, many of which are reminiscent of neural features and symptoms associated with schizophrenia. cKO mice exhibit enlarged ventricles and impaired social behaviour, locomotor activity, and learning and memory. Loss of Crmp2 in the hippocampus leads to reduced long-term potentiation, abnormal NMDA receptor composition, aberrant dendrite development and defective synapse formation in CA1 neurons. Furthermore, knockdown of crmp2 specifically in newborn neurons results in stage-dependent defects in their development during adult hippocampal neurogenesis. Our findings reveal a critical role for CRMP2 in neuronal plasticity, neural function and behavioural modulation in mice.


Subject(s)
CA1 Region, Hippocampal/metabolism , Intercellular Signaling Peptides and Proteins/deficiency , Memory Disorders/genetics , Nerve Tissue Proteins/deficiency , Neurogenesis/genetics , Neurons/metabolism , Schizophrenia/genetics , Social Behavior Disorders/genetics , Animals , CA1 Region, Hippocampal/growth & development , CA1 Region, Hippocampal/pathology , Cerebral Ventricles/growth & development , Cerebral Ventricles/metabolism , Cerebral Ventricles/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/genetics , Locomotion/genetics , Long-Term Potentiation/genetics , Male , Maze Learning , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Neurons/pathology , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/metabolism , Schizophrenia/pathology , Social Behavior Disorders/metabolism , Social Behavior Disorders/pathology , Synapses/genetics , Synapses/pathology
15.
Neurogenesis (Austin) ; 2(1): e1081715, 2015.
Article in English | MEDLINE | ID: mdl-27606328

ABSTRACT

A large number of susceptibility genes have been implicated in psychiatric disorders with a developmental origin, yet their biological roles and signaling mechanisms in neurodevelopment are largely unknown. Disrupted-In-Schizophrenia 1 (DISC1), a susceptibility gene for several major psychiatric disorders, regulates the development of newborn neurons in the adult hippocampus. Systemic pharmacological inhibition of mTOR signaling with rapamycin has been shown to rescue DISC1 deficiency-induced neurodevelopmental defects, as well as cognitive and affective deficits. Whether mTOR signaling plays a cell-autonomous and/or non-cell-autonomous role in DISC1-dependent regulation of neuronal development is not clear. Here we provide genetic evidence that hyper-activation of mTOR activator Rheb1 (Ras homolog enriched in brain 1) in newborn neurons recapitulates DISC1 deficiency-induced neurodevelopmental defects, including neuronal morphogenesis and migration. We further show that genetic deletion of Rheb1 rescues those defects in a cell-autonomous fashion in developing newborn neurons in the adult hippocampus. Our genetic and functional studies demonstrate that Rheb1 acts as a key mediator of DISC1-dependent regulation of mTOR signaling and neuronal development during adult hippocampal neurogenesis.

16.
Front Syst Neurosci ; 9: 93, 2015.
Article in English | MEDLINE | ID: mdl-26161071

ABSTRACT

Adult hippocampal neurogenesis, the constitutive generation of new granule cells in the dentate gyrus of the mature brain, is a robust model of neural development and its dysregulation has been implicated in the pathogenesis of psychiatric and neurological disorders. Previous studies in mice have shown that altered expression of Disrupted-In-Schizophrenia 1 (Disc1), the mouse homolog of a risk gene for major psychiatric disorders, results in several distinct morphological phenotypes during neuronal development. Although there are advantages to using rats over mice for neurophysiological studies, genetic manipulations have not been widely utilized in rat models. Here, we used a retroviral-mediated approach to knockdown DISC1 expression in dividing cells in the rat dentate gyrus and characterized the morphological development of adult-born granule neurons. Consistent with earlier findings in mice, we show that DISC1 knockdown in adult-born dentate granule cells in rats resulted in accelerated dendritic growth, soma hypertrophy, ectopic dendrites, and mispositioning of new granule cells due to overextended migration. Our study thus demonstrates that the Disc1 genetic manipulation approach used in prior mouse studies is feasible in rats and that there is a conserved biological function of this gene across species. Extending gene-based studies of adult hippocampal neurogenesis from mice to rats will allow for the development of additional models that may be more amenable to behavioral and in vivo electrophysiological investigations. These models, in turn, can generate additional insight into the systems-level mechanisms of how risk genes for complex psychiatric disorders may impact adult neurogenesis and hippocampal function.

17.
Neuron ; 77(4): 647-54, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-23439118

ABSTRACT

Abnormalities during brain development are thought to cause psychiatric illness and other neurodevelopmental disorders. However, developmental processes such as neurogenesis continue in restricted brain regions of adults, and disruptions of these processes could contribute to the phenotypes of neurodevelopmental disorders. As previously reported, we show that Disc1 knockdown specifically in adult-born dentate gyrus (DG) neurons results in increased mTOR signaling, hyperexcitability, and neuronal structure deficits. Disc1 knockdown also resulted in pronounced cognitive and affective deficits, which could be reversed when the affected DG neurons were inactivated. Importantly, reversing increases in mTOR signaling with an FDA-approved inhibitor both prevented and treated these behavioral deficits, even when associated structural deficits were not reversed. Our findings suggest that a component of the affective and cognitive phenotypes in neurodevelopmental disorders may be caused by disruptions in adult-born neurons. Consequently, treatments directed at this cell population may have a significant impact on these phenotypes.


Subject(s)
Cognition/physiology , Dentate Gyrus/cytology , Nerve Tissue Proteins/genetics , Neurons/cytology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Aging , Animals , Gene Knockdown Techniques , Mice , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Signal Transduction/genetics
18.
Cell Stem Cell ; 12(2): 215-23, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23395446

ABSTRACT

Adult neurogenesis, the process of generating mature neurons from adult neural stem cells, proceeds concurrently with ongoing neuronal circuit activity and is modulated by various physiological and pathological stimuli. The niche mechanism underlying the activity-dependent regulation of the sequential steps of adult neurogenesis remains largely unknown. Here, we report that neuronal activity decreases the expression of secreted frizzled-related protein 3 (sFRP3), a naturally secreted Wnt inhibitor highly expressed by adult dentate gyrus granule neurons. Sfrp3 deletion activates quiescent radial neural stem cells and promotes newborn neuron maturation, dendritic growth, and dendritic spine formation in the adult mouse hippocampus. Furthermore, sfrp3 reduction is essential for activity-induced adult neural progenitor proliferation and the acceleration of new neuron development. Our study identifies sFRP3 as an inhibitory niche factor from local mature dentate granule neurons that regulates multiple phases of adult hippocampal neurogenesis and suggests an interesting activity-dependent mechanism governing adult neurogenesis via the acute release of tonic inhibition.


Subject(s)
Hippocampus/cytology , Proteins/metabolism , Animals , Female , In Situ Hybridization , Mice , Neurogenesis/drug effects , Neurogenesis/genetics , Neurogenesis/physiology , Pilocarpine/pharmacology , Proteins/genetics , Real-Time Polymerase Chain Reaction
20.
Neuron ; 72(4): 559-71, 2011 Nov 17.
Article in English | MEDLINE | ID: mdl-22099459

ABSTRACT

Disrupted-in Schizophrenia 1 (DISC1), a susceptibility gene for major mental disorders, encodes a scaffold protein that has a multifaceted impact on neuronal development. How DISC1 regulates different aspects of neuronal development is not well understood. Here, we show that Fasciculation and Elongation Protein Zeta-1 (FEZ1) interacts with DISC1 to synergistically regulate dendritic growth of newborn neurons in the adult mouse hippocampus, and that this pathway complements a parallel DISC1-NDEL1 interaction that regulates cell positioning and morphogenesis of newborn neurons. Furthermore, genetic association analysis of two independent cohorts of schizophrenia patients and healthy controls reveals an epistatic interaction between FEZ1 and DISC1, but not between FEZ1 and NDEL1, for risk of schizophrenia. Our findings support a model in which DISC1 regulates distinct aspects of neuronal development through its interaction with different intracellular partners and such epistasis may contribute to increased risk for schizophrenia.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis , Schizophrenia/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Animals , Case-Control Studies , Cells, Cultured , Female , Gene Knockdown Techniques , Genetic Association Studies , Hippocampus/growth & development , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Polymorphism, Single Nucleotide/genetics , Protein Binding/genetics , Risk Factors , Schizophrenia/genetics
SELECTION OF CITATIONS
SEARCH DETAIL