Velo-cardio-facial syndrome/DiGeorge syndrome (VCFS/DGS) is a congenital anomaly disorder associated with hemizygous 1.5-3 Mb 22ql 1 deletions. Most patients have learning disabilities, craniofacial anomalies, outflow tract heart defects and ear disorders. Over 24 genes lie in the 1.5 Mb interval that is deleted. The 1.5 Mb region is conserved on mouse chromosome 16. By taking genetics approaches, we recently found that one of the genes, termed Tbxl, a member of the T-box family of transcription factor genes, is a strong candidate for the syndrome in mouse models. We targeted the Tbxl gene for inactivation and found that while hemizygous mice were mildly affected, homozygous mice died in the perinatal period with malformations that were particularly striking in their magnitude. They had cleft palate, major cardiovascular defects, no thymus or parathyroid glands and no outer, middle or inner ear. We are interested in determining the role of Tbxl in ear development and disease. Both the otic vesicle epithelium and surrounding periotic mesenchyme interact to form the inner ear. The fact that Tbxl is highly expressed in both tissues, suggests that it might play dual roles in patterning the inner ear. We hypothesize that Tbxl encodes a transcription factor whose dual expression is required for ear development.
For Specific Aim 1, we will determine the role of Tbxl in the otic vesicle epithelium and separately in the periotic mesenchyme by generating conditional alleles in the mouse.
For Specific Aim 2, we will determine whether Tbxl is a transcriptional activator or repressor and define the Tbxl domains required for transcription factor activity. Two other T-box genes, Tbx2 and Tbx3, are co-expressed in the otic vesicle during development. The basis for Tbxl function may lie in its requirement to form functional homodimers or heterodimers. We will determine whether Tbxl protein can functionally interact with Tbx2 or Tbx3. Many genes required to form the ear have been identified. We will take a candidate gene approach to determine the role of Tbxl in altering the expression of downstream target genes in the ears of mutant mice as Specific Aim 3. By achieving the goals of these specific aims, we will understand the role of Tbxl in normal ear development and disease. ? ?

Agency
National Institute of Health (NIH)
Institute
National Institute on Deafness and Other Communication Disorders (NIDCD)
Type
Research Project (R01)
Project #
1R01DC005186-01A1
Application #
6575431
Study Section
Special Emphasis Panel (ZRG1-IFCN-4 (05))
Program Officer
Watson, Bracie
Project Start
2002-12-01
Project End
2007-11-30
Budget Start
2002-12-01
Budget End
2003-11-30
Support Year
1
Fiscal Year
2003
Total Cost
$352,172
Indirect Cost
Name
Albert Einstein College of Medicine
Department
Genetics
Type
Schools of Medicine
DUNS #
071036636
City
Bronx
State
NY
Country
United States
Zip Code
10461
Morrow, Bernice E; McDonald-McGinn, Donna M; Emanuel, Beverly S et al. (2018) Molecular genetics of 22q11.2 deletion syndrome. Am J Med Genet A 176:2070-2081
Macchiarulo, Stephania; Morrow, Bernice E (2017) Tbx1 and Jag1 act in concert to modulate the fate of neurosensory cells of the mouse otic vesicle. Biol Open 6:1472-1482
Chung, Jonathan H; Cai, Jinlu; Suskin, Barrie G et al. (2015) Whole-Genome Sequencing and Integrative Genomic Analysis Approach on Two 22q11.2 Deletion Syndrome Family Trios for Genotype to Phenotype Correlations. Hum Mutat 36:797-807
Castellanos, Raquel; Xie, Qing; Zheng, Deyou et al. (2014) Mammalian TBX1 preferentially binds and regulates downstream targets via a tandem T-site repeat. PLoS One 9:e95151
Freyer, Laina; Nowotschin, Sonja; Pirity, Melinda K et al. (2013) Conditional and constitutive expression of a Tbx1-GFP fusion protein in mice. BMC Dev Biol 13:33
Simrick, Subreena; Szumska, Dorota; Gardiner, Jennifer R et al. (2012) Biallelic expression of Tbx1 protects the embryo from developmental defects caused by increased receptor tyrosine kinase signaling. Dev Dyn 241:1310-24
Monks, Dennis C; Morrow, Bernice E (2012) Identification of putative retinoic acid target genes downstream of mesenchymal Tbx1 during inner ear development. Dev Dyn 241:563-73
Freyer, Laina; Aggarwal, Vimla; Morrow, Bernice E (2011) Dual embryonic origin of the mammalian otic vesicle forming the inner ear. Development 138:5403-14
Freyer, Laina; Morrow, Bernice E (2010) Canonical Wnt signaling modulates Tbx1, Eya1, and Six1 expression, restricting neurogenesis in the otic vesicle. Dev Dyn 239:1708-22
Aggarwal, Vimla S; Carpenter, Courtney; Freyer, Laina et al. (2010) Mesodermal Tbx1 is required for patterning the proximal mandible in mice. Dev Biol 344:669-81

Showing the most recent 10 out of 21 publications