Rett syndrome [RTT, MIM 312750] is an X-linked postnatal developmental disorder characterized by loss of acquired skills, impairment of cognitive and motor functions, autonomic dysfunction, ataxia, tremors, seizures, autistic features, and stereotypic hand movements. RTT is caused by mutations in the X-linked MECP2 gene which encodes methyl-CpG-binding protein 2 (MeCP2). MeCP2 is thought to be a transcriptional repressor that links DNA methylation to chromatin modifications. Recently, we also found that MeCP2 interacts with an RNA-binding protein and can affect RNA splicing. We and others have shown that mutations in MECP2 cause a broad spectrum of disorders that display partial features of RTT such as autism, mild mental retardation, movement abnormalities, or seizures. Favorable X-chromosome-inactivation patterns in some of these patients and in partially symptomatic female mice that model RTT led us to propose that the diverse phenotypes of classic RTT result from MeCP2 dysfunction in a particular subset of neurons. Furthermore, we propose that loss of function of MeCP2 in specific neurons causes gene expression and RNA splicing changes that mediate the neuron-specific phenotypes. Lastly, we propose that pharmacologic therapies that target some of the neuron-specific changes linked to distinct phenotypes are likely to modulate some of these RTT phenotypes.
The Specific Aims of this project are: (1) To identify the neuroanatomical bases of several key features of RTT by deleting Mecp2 in distinct neuronal populations -- using Cre/LoxP technology-- and characterizing the phenotypes of the conditional mutant mice. (2) To identify MeCP2 targets by evaluating neuron-specific gene expression and splicing pattern changes in Mecp 308/y mice which reproduce RTT phenotypes;we will use a novel approach that employs neuron- specific BACarray lines and new splicing/expression arrays. (3) To conduct preclinical pharmacologic trials targeted at clinically relevant molecular changes (based on Data from Aims 1 and 2) to determine if such therapies will alter the RTT disease course. As a proof of concept, we will use drugs that modulate CRH and AVP activities given our discovery of the potential roles of these two MeCP2 targets in RTT phenotypes. These studies will provide insight about the neuronal subtypes and neurotransmitter systems that mediate certain key features of RTT and related disorders such as autism and X-linked mental retardation. They will also provide the community with a rich resource of neuron-specific gene expression/splicing patterns and how some of these patterns differ in RTT. Last but not least, the data generated under this study have the potential to identify effective pharmacologic interventions that could benefit RTT patients.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS057819-05
Application #
7825406
Study Section
Developmental Brain Disorders Study Section (DBD)
Program Officer
Mamounas, Laura
Project Start
2006-09-04
Project End
2011-08-31
Budget Start
2010-06-01
Budget End
2011-08-31
Support Year
5
Fiscal Year
2010
Total Cost
$360,485
Indirect Cost
Name
Baylor College of Medicine
Department
Genetics
Type
Schools of Medicine
DUNS #
051113330
City
Houston
State
TX
Country
United States
Zip Code
77030
Raman, Ayush T; Pohodich, Amy E; Wan, Ying-Wooi et al. (2018) Apparent bias toward long gene misregulation in MeCP2 syndromes disappears after controlling for baseline variations. Nat Commun 9:3225
Kee, Sara E; Mou, Xiang; Zoghbi, Huda Y et al. (2018) Impaired spatial memory codes in a mouse model of Rett syndrome. Elife 7:
Pohodich, Amy E; Yalamanchili, Hari; Raman, Ayush T et al. (2018) Forniceal deep brain stimulation induces gene expression and splicing changes that promote neurogenesis and plasticity. Elife 7:
Ito-Ishida, Aya; Yamalanchili, Hari Krishna; Shao, Yingyao et al. (2018) Genome-wide distribution of linker histone H1.0 is independent of MeCP2. Nat Neurosci 21:794-798
Lombardi, Laura M; Zaghlula, Manar; Sztainberg, Yehezkel et al. (2017) An RNA interference screen identifies druggable regulators of MeCP2 stability. Sci Transl Med 9:
Ure, Kerstin; Lu, Hui; Wang, Wei et al. (2016) Restoration of Mecp2 expression in GABAergic neurons is sufficient to rescue multiple disease features in a mouse model of Rett syndrome. Elife 5:
Meng, Xiangling; Wang, Wei; Lu, Hui et al. (2016) Manipulations of MeCP2 in glutamatergic neurons highlight their contributions to Rett and other neurological disorders. Elife 5:
Lu, Hui; Ash, Ryan T; He, Lingjie et al. (2016) Loss and Gain of MeCP2 Cause Similar Hippocampal Circuit Dysfunction that Is Rescued by Deep Brain Stimulation in a Rett Syndrome Mouse Model. Neuron 91:739-747
Zoghbi, Huda Y; Beaudet, Arthur L (2016) Epigenetics and Human Disease. Cold Spring Harb Perspect Biol 8:a019497
Sztainberg, Yehezkel; Zoghbi, Huda Y (2016) Lessons learned from studying syndromic autism spectrum disorders. Nat Neurosci 19:1408-1417

Showing the most recent 10 out of 41 publications