Elsevier

Neurobiology of Disease

Volume 35, Issue 2, August 2009, Pages 219-233
Neurobiology of Disease

Dendritic spine pathologies in hippocampal pyramidal neurons from Rett syndrome brain and after expression of Rett-associated MECP2 mutations

https://doi.org/10.1016/j.nbd.2009.05.001Get rights and content

Abstract

Rett syndrome (RTT) is an X chromosome-linked neurodevelopmental disorder associated with the characteristic neuropathology of dendritic spines common in diseases presenting with mental retardation (MR). Here, we present the first quantitative analyses of dendritic spine density in postmortem brain tissue from female RTT individuals, which revealed that hippocampal CA1 pyramidal neurons have lower spine density than age-matched non-MR female control individuals. The majority of RTT individuals carry mutations in MECP2, the gene coding for a methylated DNA-binding transcriptional regulator. While altered synaptic transmission and plasticity has been demonstrated in Mecp2-deficient mouse models of RTT, observations regarding dendritic spine density and morphology have produced varied results. We investigated the consequences of MeCP2 dysfunction on dendritic spine structure by overexpressing (∼ twofold) MeCP2-GFP constructs encoding either the wildtype (WT) protein, or missense mutations commonly found in RTT individuals. Pyramidal neurons within hippocampal slice cultures transfected with either WT or mutant MECP2 (either R106W or T158M) showed a significant reduction in total spine density after 48 h of expression. Interestingly, spine density in neurons expressing WT MECP2 for 96 h was comparable to that in control neurons, while neurons expressing mutant MECP2 continued to have lower spine density than controls after 96 h of expression. Knockdown of endogenous Mecp2 with a specific small hairpin interference RNA (shRNA) also reduced dendritic spine density, but only after 96 h of expression. On the other hand, the consequences of manipulating MeCP2 levels for dendritic complexity in CA3 pyramidal neurons were only minor. Together, these results demonstrate reduced dendritic spine density in hippocampal pyramidal neurons from RTT patients, a distinct dendritic phenotype also found in neurons expressing RTT-associated MECP2 mutations or after shRNA-mediated endogenous Mecp2 knockdown, suggesting that this phenotype represent a cell-autonomous consequence of MeCP2 dysfunction.

Introduction

Deficits in dendritic architecture are common in disorders associated with mental retardation (MR), ranging from environmental (e.g. fetal alcohol syndrome) to autosomal (e.g. Down syndrome) and X chromosome-linked forms of MR (e.g. Fragile-X, Rett syndrome; reviewed by Fiala et al., 2002, Kaufmann and Moser, 2000). A series of groundbreaking studies published in the 1970s established the precedent of abnormalities in dendritic organization in cortical neurons from humans with MR (Huttenlocher, 1970, Huttenlocher, 1974, Marin-Padilla, 1972, 1976). These series of observations consistently described reductions in the density of dendritic spines, the postsynaptic compartment of excitatory synapses (reviewed by Peters et al., 1991). In addition, a prevalence of long and tortuous spines, thought to represent immature synapses, were also commonly observed in MR and combined, these dendritic spine anomalies were referred to as “spine dysgenesis” (Purpura, 1974). Since dendritic development is a process where the formation and maturation of spines are the result of interactions between intrinsic genetic factors and external environment, the study of spine development and maintenance in MR has significant clinical relevance.

Rett syndrome (RTT) is an X chromosome-linked mental retardation that results from sporadic mutations in the gene coding for the methylated DNA-binding transcription factor MeCP2 (Amir et al., 1999; reviewed by Percy, 2001). RTT affects approximately 1:10,000 females worldwide, and prominent symptoms include deceleration of body and head growth rate, hand stereotypies and regression in motor and speech capabilities, irregularities in motor activity and breathing patterns, in addition to cognitive impairments characteristic of an autism-spectrum disorder (reviewed by Percy and Lane, 2005). Although RTT occurs predominantly in females, mutations of MECP2 have also been identified in males, where the phenotypes range from severe encephalopathy, to classic RTT, to non-specific MR (Couvert et al., 2001, Kankirawatana et al., 2006, Masuyama et al., 2005, Moog et al., 2003). While mutations of MECP2 have been associated with RTT, duplications in the chromosomal region where MECP2 is located were shown to be related to neurological disorders associated with MR, suggesting that MECP2 is a critical dosage-sensitive gene (del Gaudio et al., 2006, Smyk et al., 2008).

Morphological studies in postmortem brain samples from RTT individuals described a characteristic neuropathology, which included decreased neuronal size and increased neuronal density in the cerebral cortex, hypothalamus and the hippocampal formation (Bauman et al., 1995a,b); decreased dendritic growth in pyramidal neurons of the subiculum and frontal and motor cortices (Armstrong et al., 1995); as well as the characteristic MR-associated spine dysgenesis, observed in pyramidal neurons of the motor cortex with regions of dendrites devoid of spines (Belichenko et al., 1994). The reduction in dendritic area together with the marked decrease in dendritic spine density strongly suggests that impaired synaptic transmission is a likely pathogenic consequence of MECP2 mutations causing RTT. Indeed, the increase in neuronal expression of MECP2/Mecp2 during early brain development suggests the importance of this protein in synapse formation and maintenance (Akbarian et al., 2001, Cassel et al., 2004, Jung et al., 2003, Kaufmann et al., 2005, Mullaney et al., 2004, Shahbazian et al., 2002b). While hippocampal and cortical synaptic dysfunction in Mecp2-based mouse models of RTT has been extensively studied, observations regarding neuronal, dendritic and synaptic pathologies have produced varied results (Asaka et al., 2006, Chao et al., 2007, Dani et al., 2005, Fukuda et al., 2005, Gemelli et al., 2006, Jugloff et al., 2005, Kishi and Macklis, 2004, Moretti et al., 2006, Nelson et al., 2006, Smrt et al., 2007, Zhou et al., 2006). Here, we present the first quantitative analyses of dendritic spine density in postmortem brain tissue from RTT individuals. To identify the consequences of cell-autonomous MeCP2 dysfunction on the morphology of dendrites and dendritic spines in hippocampal pyramidal neurons, we transfected organotypic slice cultures with either wildtype or RTT-associated MECP2 mutations.

Section snippets

Human postmortem brain

All procedures on human postmortem brain samples followed national and international ethics guidelines, and were reviewed and approved by the Institutional Review Board (IRB) at The University of Alabama at Birmingham (UAB). Brain sections containing the hippocampal formation were obtained from individuals diagnosed with RTT, and unaffected (non-MR) individuals served as controls. Postmortem human tissue was obtained from the NICHD Brain and Tissue Bank for Developmental Disorders at the

Hippocampal CA1 pyramidal neurons from RTT individuals have lower dendritic spine density than those from non-MR individuals

The only prior study on the features of dendritic spines in RTT individuals was performed on cortical samples and lacked quantitative statistical analyses (Belichenko et al., 1994). To perform quantitative analyses of spine density in pyramidal neurons from human hippocampus, postmortem tissue was obtained from female RTT individuals that were 1 to 42 years of age, and compared to age-matched unaffected non-MR female individuals (see Table 1 for all available information from the human brain

Discussion

Here, we present several novel observations regarding dendritic spine dysgenesis in RTT individuals and the role of the transcriptional regulator MeCP2 on dendritic complexity and dendritic spine density and morphology. First, we show that CA1 pyramidal neurons of the hippocampus in female individuals with RTT have lower dendritic spine density than age-matched unaffected (non-MR) female individuals, as observed qualitatively in pyramidal neurons of the motor cortex (Belichenko et al., 1994).

Acknowledgments

Supported by NIH grants NS40593 and NS057780, IRSF and the Civitan International Foundation (LP-M). We also thank the assistance of the UAB Intellectual and Developmental Disabilities Research Center (IDDRC; P30-HD38985) and the UAB Neuroscience Cores (P30-NS47466, P30-NS57098). Human tissue was obtained from the NICHD Brain and Tissue Bank for Developmental Disorders at the University of Maryland, Baltimore, MD, and the Harvard Brain Bank. We thank Dr. Carolyn Schanen (Nemours Biomedical

References (81)

  • GemelliT. et al.

    Postnatal loss of methyl-CpG binding protein 2 in the forebrain is sufficient to mediate behavioral aspects of Rett syndrome in mice

    Biol. Psychiatry

    (2006)
  • GhoshR.P. et al.

    Rett syndrome-causing mutations in human MeCP2 result in diverse structural changes that impact folding and DNA interactions

    J. Biol. Chem.

    (2008)
  • HarrisK.M.

    Structure, development, and plasticity of dendritic spines

    Curr. Opin. Neurobiol.

    (1999)
  • HoK.L. et al.

    MeCP2 binding to DNA depends upon hydration at methyl-CpG

    Mol. Cell

    (2008)
  • JugloffD.G. et al.

    Increased dendritic complexity and axonal length in cultured mouse cortical neurons overexpressing methyl-CpG-binding protein MeCP2

    Neurobiol. Dis.

    (2005)
  • KasaiH. et al.

    Structure–stability–function relationships of dendritic spines

    Trends Neurosci.

    (2003)
  • KaufmannW.E. et al.

    Cyclooxygenase-2 expression during rat neocortical development and in Rett syndrome

    Brain Dev.

    (1997)
  • KaufmannW.E. et al.

    MeCP2 expression and function during brain development: implications for Rett syndrome's pathogenesis and clinical evolution

    Brain Dev.

    (2005)
  • KishiN. et al.

    MECP2 is progressively expressed in post-migratory neurons and is involved in neuronal maturation rather than cell fate decisions

    Mol. Cell. Neurosci.

    (2004)
  • KudoS. et al.

    Functional analyses of MeCP2 mutations associated with Rett syndrome using transient expression systems

    Brain Dev.

    (2001)
  • LarimoreJ. et al.

    Bdnf overexpression in hippocampal neurons prevents dendritic atrophy caused by Rett-associated MECP2 mutations

    Neurobiol. Dis.

    (2009)
  • Marin-PadillaM.

    Structural abnormalities of the cerebral cortex in human chromosomal aberrations: a Golgi study

    Brain Res.

    (1972)
  • MasuyamaT. et al.

    Classic Rett syndrome in a boy with R133C mutation of MECP2

    Brain Dev.

    (2005)
  • MoogU. et al.

    Neurodevelopmental disorders in males related to the gene causing Rett syndrome in females (MECP2)

    Eur. J. Paediatr. Neurol.

    (2003)
  • MullaneyB.C. et al.

    Developmental expression of methyl-CpG binding protein 2 is dynamically regulated in the rodent brain

    Neuroscience

    (2004)
  • NelsonE.D. et al.

    MeCP2-dependent transcriptional repression regulates excitatory neurotransmission

    Curr. Biol.

    (2006)
  • PercyA.K.

    Rett syndrome: clinical correlates of the newly discovered gene

    Brain. Dev.

    (2001)
  • ShahbazianM. et al.

    Mice with truncated MeCP2 recapitulate many Rett syndrome features and display hyperacetylation of histone H3

    Neuron

    (2002)
  • SmrtR.D. et al.

    Mecp2 deficiency leads to delayed maturation and altered gene expression in hippocampal neurons

    Neurobiol. Dis.

    (2007)
  • StoppiniL. et al.

    A simple method for organotypic cultures of nervous tissue

    J. Neurosci. Methods

    (1991)
  • ZhouQ. et al.

    Shrinkage of dendritic spines associated with long-term depression of hippocampal synapses

    Neuron

    (2004)
  • ZhouZ. et al.

    Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation

    Neuron

    (2006)
  • AlonsoM. et al.

    ERK1/2 activation is necessary for BDNF to increase dendritic spine density in hippocampal CA1 pyramidal neurons

    Learn. Mem.

    (2004)
  • AmirR.E. et al.

    Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2

    Nat. Genet.

    (1999)
  • ArmstrongD. et al.

    Selective dendritic alterations in the cortex of Rett syndrome

    J. Neuropathol. Exp. Neurol.

    (1995)
  • BallestarE. et al.

    Effects of Rett syndrome mutations of the methyl-CpG binding domain of the transcriptional repressor MeCP2 on selectivity for association with methylated DNA

    Biochemistry

    (2000)
  • BastrikovaN. et al.

    Synapse elimination accompanies functional plasticity in hippocampal neurons

    Proc. Natl. Acad. Sci. U. S. A.

    (2008)
  • BaumanM.L. et al.

    Microscopic observations of the brain in Rett syndrome

    Neuropediatrics

    (1995)
  • BaumanM.L. et al.

    Pervasive neuroanatomic abnormalities of the brain in three cases of Rett's syndrome

    Neurology

    (1995)
  • BelichenkoP.V. et al.

    Rett syndrome: 3-D confocal microscopy of cortical pyramidal dendrites and afferents

    NeuroReport

    (1994)
  • Cited by (195)

    • iPSC-derived models of autism: Tools for patient phenotyping and assay-based drug discovery

      2022, Phenotyping of Human iPSC-derived Neurons: Patient-Driven Research
    • Rett syndrome: from the involved gene(s) to treatment

      2022, Neurobiology of Brain Disorders: Biological Basis of Neurological and Psychiatric Disorders, Second Edition
    • Dendritic spine membrane proteome and its alterations in autistic spectrum disorder

      2022, Advances in Protein Chemistry and Structural Biology
    View all citing articles on Scopus
    View full text