Breathing disorders in Rett syndrome: Progressive neurochemical dysfunction in the respiratory network after birth

https://doi.org/10.1016/j.resp.2009.04.017Get rights and content

Abstract

Disorders of respiratory control are a prominent feature of Rett syndrome (RTT), a severely debilitating condition caused by mutations in the gene encoding methyl-CpG-binding protein 2 (MECP2). RTT patients present with a complex respiratory phenotype that can include periods of hyperventilation, apnea, breath holds terminated by Valsalva maneuvers, forced and deep breathing and apneustic breathing, as well as abnormalities of heart rate control and cardiorespiratory integration. Recent studies of mouse models of RTT have begun to shed light on neurologic deficits that likely contribute to respiratory dysfunction including, in particular, defects in neurochemical signaling resulting from abnormal patterns of neurotransmitter and neuromodulator expression. The authors hypothesize that breathing dysregulation in RTT results from disturbances in mechanisms that modulate the respiratory rhythm, acting either alone or in combination with more subtle disturbances in rhythm and pattern generation. This article reviews the evidence underlying this hypothesis as well as recent efforts to translate our emerging understanding of neurochemical defects in mouse models of RTT into preclinical trials of potential treatments for respiratory dysfunction in this disease.

Introduction

Rett syndrome (RTT) is a complex neurodevelopmental disorder whose underlying pathogenic mechanisms remain poorly understood. RTT affects approximately 1 in 10,000 live female births and is characterized by apparently normal early postnatal development followed by neurological decline around 6–18 months of age. The disorder has a highly variable course and affected individuals exhibit a broad array of symptoms that generally includes loss of acquired speech, head growth deceleration, autistic features such as emotional withdrawal and diminished eye contact, motor stereotypies, early hypotonia followed by rigidity, epileptiform seizures, exaggerated responses to stress and severe respiratory and autonomic (cardiac and gastrointestinal) dysfunction (Hagberg et al., 1983, Shahbazian et al., 2002, Vorsanova et al., 2004, Chahrour and Zoghbi, 2007, Chahrour et al., 2008, Ogier and Katz, 2008, Weese-Mayer et al., 2006, Weese-Mayer et al., 2008). Approximately 25% of RTT patients may die prematurely of cardiorespiratory failure (Kerr et al., 1997).

At least 95% of typical RTT cases result from loss-of-function mutations in the gene encoding methyl-CpG-binding protein 2 (MeCP2; Amir et al., 1999, Shahbazian et al., 2002), one of a number of methyl-binding proteins (Klose and Bird, 2006) that regulate gene expression by repressing transcription at methylated promoters. Over 200 different MECP2 mutations have been found in RTT patients and tend to cluster within two functional domains of the protein; a methyl-binding domain that recognizes methylated CpG dinucleotides with particular flanking sequences (Klose and Bird, 2006), and a transcription repression domain. The MECP2 gene is X-linked, and homozygous mutation in females (or hemizygous mutation in males) is invariably lethal. Thus, affected females are heterozygotes and somatic mosaics for MeCP2, i.e., cells in which the mutated allele occurs on the inactive X are phenotypically normal, whereas cells in which the mutated allele occurs on the active X are mutant. Disease phenotype is therefore affected not only by the specific MECP2 mutation but by the skewing of X chromosome inactivation; individuals in which inactivation is skewed towards the mutant allele are less severely affected, and vice versa. For a more detailed discussion of molecular genetic aspects of RTT the reader is referred to an excellent recent review by Chahrour and Zoghbi (2007).

Section snippets

Cardiorespiratory phenotypes in RTT

Breathing abnormalities are among the clinical diagnostic criteria for RTT, and include alternating periods of hyperventilation and apneas, breath holds terminated by Valsalva's maneuvers, forced and deep breathing as well as apneustic breathing (Fig. 1) (cf. Elian and Rudolf, 1991, Julu et al., 2001, Julu and Witt Engerström, 2005, Marcus et al., 1994, Weese-Mayer et al., 2006, Weese-Mayer et al., 2008). Perhaps the most striking breathing abnormalities are the frequent breath holds and apneas

Respiratory phenotypes in mouse models of RTT

The discovery that mutations in MECP2 are responsible for most cases of RTT led to the development of mouse models in which Mecp2 is either deleted, mutated or overexpressed, including (1) Mecp2−/y mice with extended exonic deletion of the Mecp2 gene (Chen et al., 2001, Guy et al., 2001, Pelka et al., 2006), (2) Mecp2308/y mice with truncation of MeCP2 protein at amino acid 308, a human RTT mutation (Shahbazian et al., 2002), (3) Mecp2Flox/y mice expressing a hypomorphic Mecp2 allele (Samaco et

Disturbed GABAergic and glutamatergic neurotransmission

Growing evidence suggests that at least some of the neurological endophenotypes in RTT, including breathing abnormalities, are associated with changes in MeCP2-mediated regulation of excitatory glutamatergic or inhibitory GABAergic synaptic transmission (Blue et al., 1999, Samaco et al., 2005, Chao et al., 2007, Stettner et al., 2007). Moreover, significant metabolic anomalies, including altered glutamate metabolism, have been described in Mecp2 null mice (Viola et al., 2007). It is well

Experimental strategies to treat breathing disorders in Mecp2 null mice

Recent studies in conditional Mecp2 null mice have demonstrated that reactivation of the Mecp2 gene, even in severely symptomatic animals, can rescue neurologic function to a remarkable degree (Guy et al., 2007; see also Giacometti et al., 2007). These findings indicate that deficits caused by loss of MeCP2 function reflect defects in neuronal signaling, rather than neurodegeneration, and, therefore, are potentially reversible. In addition, genetic overexpression of the BDNF gene in Mecp2

Conclusions

Our current understanding of the respiratory phenotype in RTT leads us to the following conclusions:

  • 1.

    The RTT breathing phenotype is extremely complex (hyperventilation, breath holds, forced and deep breathing and apneustic breathing) and includes disturbances in cardiorespiratory coupling. This complexity suggests that cardiorespiratory dysfunction in RTT is unlikely to result from a single underlying mechanism.

  • 2.

    The RTT breathing phenotype is abnormal during sleep and wakefulness; however, the

Summary

The complexity of the RTT breathing phenotype may well be related to the diversity of neurochemical changes caused by loss of MeCP2 function, as all of the affected neurotransmitter and neuromodulatory systems described thus far are important for expression, modulation and/or adaptation of respiratory motor output. Further advances in understanding cardiorespiratory pathophysiology in RTT will benefit from integrative experimental strategies that are able to link specific neurochemical deficits

Acknowledgements

The authors gratefully acknowledge funding support from the following organizations: DMK (United States National Institutes of Health/National Institute of Neurological Diseases and Stroke, Rett Syndrome Research Foundation (RSRF) and the International Rett Syndrome Foundation (IRSF)); MD (IRSRF and the Deutsche Forschungsgemeinschaft (DFG) Research Center for Molecular Physiology of the Brain (CMPB Göttingen/Germany)); JMR (United States National Institutes of Health (NIH)/National Heart,

References (110)

  • M. Dutschmann et al.

    Postnatal emergence of synaptic plasticity associated with dynamic adaptation of the respiratory motor pattern

    Respir. Physiol. Neurobiol.

    (2008)
  • L.H. Gargaglioni et al.

    Brain monoaminergic neurons and ventilatory control in vertebrates

    Respir. Physiol. Neurobiol.

    (2008)
  • C. Gestreau et al.

    Activation of XII motoneurons and premotor neurons during various oropharyngeal behaviors

    Respir. Physiol. Neurobiol.

    (2005)
  • G. Hilaire

    Endogenous noradrenaline affects the maturation and function of the respiratory network: possible implication for SIDS

    Auton. Neurosci.

    (2006)
  • G. Hilaire et al.

    Modulation of the respiratory rhythm generator by the pontine noradrenergic A5 and A6 groups in rodents

    Respir. Physiol. Neurobiol.

    (2004)
  • S. Ide et al.

    Defect in normal developmental increase of the brain biogenic amine concentrations in the Mecp2-null mouse

    Neurosci. Lett.

    (2005)
  • M. Ingvar et al.

    Enhancement by an ampakine of memory encoding in humans

    Exp. Neurol.

    (1997)
  • P.O. Julu et al.

    Assessment of the maturity-related brainstem functions reveals the heterogeneous phenotypes and facilitates clinical management of Rett syndrome

    Brain Dev.

    (2005)
  • D.M. Katz

    Neurotrophic and transcriptional regulation of respiratory neuron development

    Resp. Physiol. Neurobiol.

    (2005)
  • D.D. Kline et al.

    Mecp2 null mice exhibit enhanced excitatory postsynaptic currents (EPSCs) in the brainstem nucleus tractus solitarius (NTS)

    Soc. Neurosci. Abstr.

    (2008)
  • R.J. Klose et al.

    Genomic DNA methylation: the mark and its mediators

    Trends Biochem. Sci.

    (2006)
  • S. Lee et al.

    Activity-dependent NR2B expression is mediated by MeCP2-dependent epigenetic regulation

    Biochem. Biophys. Res. Commun.

    (2008)
  • A. Lekman et al.

    Rett syndrome: biogenic amines and metabolites in postmortem brain

    Pediatr. Neurol.

    (1989)
  • G. Lynch

    Glutamate-based therapeutic approaches: ampakines

    Curr. Opin. Pharmacol.

    (2006)
  • C.L. Marcus et al.

    Polysomnographic characteristics of patients with Rett syndrome

    J. Pediatr.

    (1994)
  • L.M. Monteggia et al.

    Rett syndrome and the impact of MeCP2 associated transcriptional mechanisms on neurotransmission

    Biol. Psychiatry

    (2009)
  • J.B. Nielsen et al.

    Biochemical and clinical effects of tyrosine and tryptophan in the Rett syndrome

    Brain Dev.

    (1990)
  • Y. Nomura et al.

    Rett syndrome—an early catecholamine and indolamine deficient disorder?

    Brain Dev.

    (1985)
  • M. Ogier et al.

    Breathing dysfunction in Rett Syndrome: understanding epigenetic regulation of the respiratory network

    Resp. Physiol. Neurobiol.

    (2008)
  • D.W. Richter et al.

    Serotonin receptors: guardians of stable breathing

    Trends Mol. Med.

    (2003)
  • P. Riederer et al.

    Neurochemical aspects of the Rett syndrome

    Brain Dev.

    (1985)
  • C.B. Saper et al.

    The sleep switch: hypothalamic control of sleep and wakefulness

    Trends Neurosci.

    (2001)
  • A. Schousboe et al.

    Role of astrocytic transport processes in glutamatergic and GABAergic neurotransmission

    Neurochem. Int.

    (2004)
  • E.A. Sekul et al.

    Electrocardiographic findings in Rett syndrome: an explanation for sudden death?

    J. Pediatr.

    (1994)
  • M. Shahbazian et al.

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

    Neuron

    (2002)
  • T. Temudo et al.

    Evaluation of CSF neurotransmitters and folate in 25 patients with Rett disorder and effects of treatment

    Brain Dev.

    (2009)
  • S. Akbarian et al.

    Brain-derived neurotrophic factor is essential for opiate-induced plasticity of noradrenergic neurons

    J. Neurosci.

    (2002)
  • R.E. Amir et al.

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

    Nat. Genet.

    (1999)
  • T.L. Baker-Herman et al.

    BDNF is necessary and sufficient for spinal respiratory plasticity following intermittent hypoxia

    Nat. Neurosci.

    (2004)
  • A. Balkowiec et al.

    Cellular mechanisms of activity-dependent release of native BDNF from hippocampal neurons

    J. Neurosci.

    (2002)
  • A. Balkowiec et al.

    Brain-derived neurotrophic factor acutely inhibits AMPA currents in developing sensory relay neurons

    J. Neurosci.

    (2000)
  • A. Balkowiec et al.

    Activity-dependent release of endogenous brain-derived neurotrophic factor from primary sensory neurons detected by ELISA in situ

    J. Neurosci.

    (2000)
  • A. Balkowiec et al.

    Brain-derived neurotrophic factor is required for normal development of the central respiratory rhythm in mice

    J. Physiol.

    (1998)
  • N. Ballas et al.

    Non-cell autonomous influence of MeCP2-deficient glia on neuronal dendritic morphology

    Nat. Neurosci.

    (2009)
  • J. Bissonnette et al.

    Possible role of bioaminergic systems in the respiratory disorders of Rett syndrome

  • J.M. Bissonnette et al.

    Separate respiratory phenotypes in methyl-CpG-binding protein 2 (Mecp2) deficient mice

    Pediatr. Res.

    (2006)
  • J.M. Bissonnette et al.

    Effect of inspired oxygen on periodic breathing in methy-CpG-binding protein 2 (Mecp2) deficient mice

    J. Appl. Physiol.

    (2008)
  • C. Bou-Flores et al.

    Abnormal phrenic motoneuron activity and morphology in neonatal monoamine oxidase A-deficient transgenic mice: possible role of a serotonin excess

    J. Neurosci.

    (2000)
  • J. Bouvier et al.

    Brain-derived neurotrophic factor enhances fetal respiratory rhythm frequency in the mouse preBötzinger complex in vitro

    Eur. J. Neurosci.

    (2008)
  • H. Bras et al.

    Prenatal activation of 5-HT2A receptor induces expression of 5-HT1B receptor in phrenic motoneurons and alters the organization of their premotor network in newborn mice

    Eur. J. Neurosci.

    (2008)
  • Cited by (145)

    • Breathing disturbances in Rett syndrome

      2022, Handbook of Clinical Neurology
    • Rett syndrome and MECP2-related disorders

      2020, Neurodevelopmental Disorders: Comprehensive Developmental Neuroscience
    • ANAVEX®2-73 (blarcamesine), a Sigma-1 receptor agonist, ameliorates neurologic impairments in a mouse model of Rett syndrome

      2019, Pharmacology Biochemistry and Behavior
      Citation Excerpt :

      In addition, enhanced pauses were also monitored as this parameter is commonly used to assess airway responsiveness or obstruction (Hamelmann et al., 1997; Zhu et al., 2011; Oldenburg et al., 2012). In the context of RTT and Mecp2 deficiency, enhanced pause provides another metric of Te, in particular the early expiration phase that appears to be more affected in Mecp2 deficient animals (Katz et al., 2009). Body weights were recorded twice weekly in both the younger and older groups of mice.

    View all citing articles on Scopus

    The following abbreviations are used for the gene encoding methyl-CpG-binding protein 2 and its respective protein product: MECP2 (human gene): Mecp2 (mouse gene), MeCP2 (protein).

    View full text