Hostname: page-component-8448b6f56d-c47g7 Total loading time: 0 Render date: 2024-04-23T07:02:10.865Z Has data issue: false hasContentIssue false

Rubinstein–Taybi syndrome: clinical and molecular overview

Published online by Cambridge University Press:  20 August 2007

Jeroen H. Roelfsema
Affiliation:
Dept of Human Genetics, Center for Human and Clinical Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
Dorien J.M. Peters*
Affiliation:
Dept of Human Genetics, Center for Human and Clinical Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
*
*Corresponding author: Dorien Peters, Dept of Human Genetics, Center for Human and Clinical Genetics, Leiden University Medical Center, Postzone S-04-P, PO Box 9600, 2300 RC Leiden, The Netherlands. Tel: 31 71 5269490; Fax: 31 71 5268285; E-mail: d.j.m.peters@lumc.nl

Abstract

Rubinstein–Taybi syndrome is characterised by mental retardation, growth retardation and a particular dysmorphology. The syndrome is rare, with a frequency of approximately one affected individual in 100 000 newborns. Mutations in two genes – CREBBP and EP300 – have been identified to cause the syndrome. These two genes show strong homology and encode histone acetyltransferases (HATs), which are transcriptional co-activators involved in many signalling pathways. Loss of HAT activity is sufficient to account for the phenomena seen in Rubinstein–Taybi patients. Although some mutations found in CREBBP are translocations, inversions and large deletions, most are point mutations or small deletions and insertions. Mutations in EP300 are comparatively rare. Extensive screening of patients has revealed mutations in CREBBP and EP300 in around 50% of cases. The cause of the syndrome in the remaining patients remains to be identified, but other genes could also be involved. Here, we describe the clinical presentation of Rubinstein–Taybi syndrome, review the mutation spectrum and discuss the current understanding of causative molecular mechanisms.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2007

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Hennekam, R.C.M. et al. (1989) Rubinstein–Taybi Syndrome in a mother and son. Eur J Pediatr 148, 439-441CrossRefGoogle Scholar
2Marion, R.W., Garcia, D.M., Karasik, J.B. (1993) Apparent dominant transmission of the Rubinstein–Taybi syndrome. Am J Med Genet 46, 284-287CrossRefGoogle ScholarPubMed
3Petrij, F. et al. (2000) Diagnostic analysis of the Rubinstein–Taybi syndrome: five cosmids should be used for microdeletion detection and low number of protein truncating mutations. J Med Genet 37, 168-76CrossRefGoogle ScholarPubMed
4Petrij, F. et al. (1995) Rubinstein–Taybi syndrome is caused by mutations in the transcriptional co-activator CBP. Nature 376, 348-351CrossRefGoogle ScholarPubMed
5Imaizumi, K. and Kuroki, Y. (1991) Rubinstein–Taybi syndrome with de novo reciprocal translocation t(2;16)(p13.3;p13.3). Am J Med Genet 38, 636-639CrossRefGoogle Scholar
6Tommerup, N., van der Hagen, C.B. and Heiberg, A. (1991) Tentative assignment of a locus for Rubinstein–Taybi Syndrome to 16p13.3 by a de novo reciprocal translocation, t(7;16)(q34;p13.3). Cytogenet Cell Genet 58, 2002Google Scholar
7Lacombe, D. et al. (1992) Confirmation of assignment of a locus for Rubinstein–Taybi syndrome gene to 16p13.3. Am J Med Genet 44, 126-128CrossRefGoogle ScholarPubMed
8Breuning, M.H. et al. (1993) Rubinstein–Taybi Syndrome caused by submicroscopic deletions of chromosome 16. Am J Hum Genet 52, 249-54Google Scholar
9Bartsch, O. et al. (2005) DNA sequencing of CREBBP demonstrates mutations in 56% of patients with Rubinstein–Taybi syndrome (RSTS) and in another patient with incomplete RSTS. Hum Genet 117, 485-493CrossRefGoogle ScholarPubMed
10Roelfsema, J.H. et al. (2005) Genetic Heterogeneity in Rubinstein–Taybi Syndrome: Mutations in Both the CBP and EP300 Genes Cause Disease. Am J Hum Genet 76, 572-580CrossRefGoogle ScholarPubMed
11Rubinstein, J.H. and Taybi, H. (1963) Broad thumbs and toes and facial abnormalities. A possible mental retardation syndrome. Am J Dis Child 105, 588-608CrossRefGoogle ScholarPubMed
12Michail, J., Matsoukas, J. and Theodorou, S. (1957) Arched, clubbed thumb in strong abduction-extension & other concomitant symptoms. Rev Chir Orthop Reparatrice Appar Mot 43, 142-146Google ScholarPubMed
13Stevens, C.A. and Bhakta, M.G. (1995) Cardiac abnormalities in the Rubinstein-Taybi syndrome. Am J Med Genet 59, 346-348CrossRefGoogle ScholarPubMed
14Hennekam, R.C.M. et al. (1990) Rubinstein–Taybi syndrome in the Netherlands. Am J Med Genet Suppl. 6, 17-29Google ScholarPubMed
15Wiley, S. (2003) Rubinstein-Taybi syndrome medical guidelines. Am J Med Genet 119, 101-110CrossRefGoogle Scholar
16Rubinstein, J.H. (1990) Broad thumb-hallux (Rubinstein–Taybi) Syndrome 1957-1988. Am J Med Genet Suppl. 6, 3-16Google ScholarPubMed
17Hennekam, R.C.M., Boogaard, M.-J. and Doorne, J.M. (1991) A cephalometric study in Rubinstein–Taybi syndrome. J Craniofac Genet Dev Biol 11, 33-40Google ScholarPubMed
18Hennekam, R.C.M. et al. (1990) Metacarpophalangeal pattern profile analysis in Rubinstein–Taybi Syndrome. Am J Med Genet Suppl. 6, 48-50Google ScholarPubMed
19Hennekam, R.C. et al. (1993) Deletion at chromosome 16p13.3 as a cause of Rubinstein-Taybi syndrome: clinical aspects. Am J Hum Genet 52, 255-262Google ScholarPubMed
20Hennekam, R.C.M. et al. (1992) Psychological and speech studies in Rubinstein–Taybi syndrome. Am J Mental Retard 96, 645-660Google ScholarPubMed
21Miller, R.W. and Rubinstein, J.H. (1995) Tumors in Rubinstein–Taybi syndrome. Am J Med Genet 56, 112-115CrossRefGoogle ScholarPubMed
22Hennekam, R.C.M., Stevens, C.A. and van de Kamp, J.J.P. (1990) Etiology and recurrence risk in Rubinstein–Taybi Syndrome. Am J Med Genet Suppl. 6, 56-64Google ScholarPubMed
23Sobel, R.A. and Woerner, S. (1981) Rubinstein–Taybi syndrome and nasopharyngeal rhabdomyosarcoma. J Pediatr 99, 1000-1001CrossRefGoogle ScholarPubMed
24Bonioli, E. and Bellini, C. (1992) Rubinstein–Taybi Syndrome and pheochromocytoma. Am J Med Genet 44, 386CrossRefGoogle ScholarPubMed
25Bilir, B.M., Bilir, N. and Wilson, G.N. (1990) Intracranial angioblastic meningioma and an aged appearance in a woman with Rubinstein–Taybi Syndrome. Am J Med Genet Suppl. 6, 69-72Google Scholar
26Cambiaghi, S. et al. (1994) Multiple pilomatricomas in Rubinstein–Taybi syndrome: a case report. Pediatr Dermatol 11, 21-25CrossRefGoogle ScholarPubMed
27Urlinger, S. et al. (2000) Exploring the sequence space for tetracycline-dependent transcriptional activators: novel mutations yield expanded range and sensitivity. Proc Natl Acad Sci U S A 97, 7963-7968CrossRefGoogle ScholarPubMed
28Russell, N.A., Hoffman, H.J. and Bain, H.W. (1971) Intraspinal neurilemoma in association with the Rubinstein–Taybi syndrome. Pediatrics 47, 444-447CrossRefGoogle ScholarPubMed
29Jonas, D.M., Heilbron, D.C. and Ablin, A.R. (1978) Rubinstein–Taybi syndrome and acute leukemia. J Pediatr. 92, 851-852CrossRefGoogle ScholarPubMed
30Goodfellow, A., Emmerson, R.W. and Calvert, H.T. (1980) Rubinstein–Taybi Syndrome and spontaneous keloids. Clin Exp Dermatol 5, 369-371CrossRefGoogle ScholarPubMed
31Rohlfing, B., Lewis, K. and Singleton, E.B. (1971) Rubinstein–Taybi syndrome. Report of an unusual case. Am J Dis Child 121, 71-74CrossRefGoogle ScholarPubMed
32Kwok, R.P. et al. (1994) Nuclear protein CBP is a coactivator for the transcription factor CREB. Nature 370, 223-226CrossRefGoogle ScholarPubMed
33Lundblad, J.R. et al. (1995) Adenoviral E1A-associated protein p300 as a functional homologue of the transcriptional co-activator CBP. Nature 374, 85-88CrossRefGoogle ScholarPubMed
34Chrivia, J.C. et al. (1993) Phosphorylated CREB binds specifically to the nuclear protein CBP. Nature 365, 855-859CrossRefGoogle Scholar
35Shi, Y. and Mello, C. (1998) A CBP/p300 homolog specifies multiple differentiation pathways in Caenorhabditis elegans. Genes Dev 12, 943-955CrossRefGoogle ScholarPubMed
36Bordoli, L. et al. (2001) Plant orthologs of p300/CBP: conservation of a core domain in metazoan p300/CBP acetyltransferase-related proteins. Nucleic Acids Res. 29, 589-597CrossRefGoogle ScholarPubMed
37Giles, R.H., Peters, D.J.M. and Breuning, M.H. (1998) Conjunction dysfunction: CBP/p300 in human disease. Trends Genet. 14, 178-183CrossRefGoogle ScholarPubMed
38Goodman, R.H. and Smolik, S. (2000) CBP/p300 in cell growth, transformation, and development. Genes Dev 14, 1553-1577CrossRefGoogle ScholarPubMed
39Bannister, A.J. and Kouzarides, T. (1996) The CBP co-activator is a histone acetyltransferase. Nature 384, 641-643CrossRefGoogle ScholarPubMed
40Ogryzko, V.V. et al. (1996) The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell 87, 953-959CrossRefGoogle ScholarPubMed
41Gu, W. and Roeder, R.G. (1997) Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 90, 595-606CrossRefGoogle ScholarPubMed
42Roth, S.Y., Denu, J.M. and Allis, C.D. (2001) Histone acetyltransferases. Annu Rev Biochem 70, 81-120CrossRefGoogle ScholarPubMed
43Kung, A.L. et al. (2000) Gene dose-dependent control of hematopoiesis and hematologic tumor suppression by CBP. Genes Dev 14, 272-277CrossRefGoogle ScholarPubMed
44Oike, Y. et al. (1999) Truncated CBP protein leads to classical Rubinstein–Taybi syndrome phenotypes in mice: implications for a dominant-negative mechanism. Hum Mol Genet 8, 387-396CrossRefGoogle ScholarPubMed
45Tanaka, Y. et al. (2000) Extensive brain hemorrhage and embryonic lethality in a mouse null mutant of CREB-binding protein. Mech Dev 95, 133-145CrossRefGoogle Scholar
46Yao, T.P. et al. (1998) Gene dosage-dependent embryonic development and proliferation defects in mice lacking the transcriptional integrator p300. Cell 93, 361-372CrossRefGoogle ScholarPubMed
47Kalkhoven, E. (2004) CBP and p300: HATs for different occasions. Biochem Pharmacol 68, 1145-1155CrossRefGoogle ScholarPubMed
48Kasper, L.H. et al. (2006) Conditional knockout mice reveal distinct functions for the global transcriptional coactivators CBP and p300 in T-cell development. Mol Cell Biol 26, 789-809CrossRefGoogle ScholarPubMed
49Kwok, R.P., Liu, X.T. and Smith, G.D. (2006) Distribution of co-activators CBP and p300 during mouse oocyte and embryo development. Mol Reprod Dev 73, 885-894CrossRefGoogle ScholarPubMed
50Partanen, A., Motoyama, J. and Hui, C.C. (1999) Developmentally regulated expression of the transcriptional cofactors/histone acetyltransferases CBP and p300 during mouse embryogenesis. Int J Dev Biol 43, 487-494Google ScholarPubMed
51Kawasaki, H. et al. (1998) Distinct roles of the co-activators p300 and CBP in retinoic-acid-induced F9-cell differentiation. Nature 393, 284-289CrossRefGoogle ScholarPubMed
52Ugai, H., Uchida, K., Kawasaki, H. and Yokoyama, K.K. (1999) The coactivators p300 and CBP have different functions during the differentiation of F9 cells. J Mol Med 77, 481-494CrossRefGoogle ScholarPubMed
53Roth, J.F. et al. (2003) Differential role of p300 and CBP acetyltransferase during myogenesis: p300 acts upstream of MyoD and Myf5. EMBO J 22, 5186-5196CrossRefGoogle ScholarPubMed
54Tanaka, Y. et al. (1997) Abnormal skeletal patterning in embryos lacking a single Cbp allele: a partial similarity with Rubinstein–Taybi syndrome. Proc Natl Acad Sci U S A 94, 10215-10220CrossRefGoogle ScholarPubMed
55Bartholdi, D. et al. (2007) Genetic heterogeneity in Rubinstein–Taybi Syndrome: delineation of the phenotype of the first patients carrying mutations in EP300. J Med Genet 44, 327-333CrossRefGoogle ScholarPubMed
56Petrij, F. et al. (2000) Rubinstein–Taybi syndrome caused by a De Novo reciprocal translocation t(2;16)(q36.3;p13.3). Am J Med Genet 92, 47-523.0.CO;2-H>CrossRefGoogle Scholar
57Giles, R.H. et al. (1997) Construction of a 1.2-Mb contig surrounding and molecular analysis of, the human CREB-binding protein (CBP/CREBBP) gene on chromosome 16p13.3. Genomics 42, 96-114CrossRefGoogle ScholarPubMed
58Troke, P.J., Kindle, K.B., Collins, H.M. and Heery, D.M. (2006) MOZ fusion proteins in acute myeloid leukaemia. Biochem Soc Symp 73, 23-39Google Scholar
59Bartsch, O. et al. (2006) Evidence for a new contiguous gene syndrome, the chromosome 16p13.3 deletion syndrome alias severe Rubinstein–Taybi syndrome. Hum Genet 120, 179-186CrossRefGoogle ScholarPubMed
60Schouten, J.P. et al. (2002) Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification. Nucleic Acids Res 30, e57CrossRefGoogle ScholarPubMed
61Bentivegna, A. et al. (2006) Rubinstein–Taybi Syndrome: spectrum of CREBBP mutations in Italian patients. BMC Med Genet 7, 77CrossRefGoogle ScholarPubMed
62Bartsch, O. et al. (1999) FISH studies in 45 patients with Rubinstein–Taybi syndrome: deletions associated with polysplenia, hypoplastic left heart and death in infancy. Eur J Human Genet 7, 748-756CrossRefGoogle ScholarPubMed
63Coupry, I. et al. (2004) Analysis of CBP (CREBBP) gene deletions in Rubinstein–Taybi syndrome patients using real-time quantitative PCR. Hum Mutat 23, 278-284CrossRefGoogle ScholarPubMed
64Udaka, T. et al. (2006) Screening for Partial Deletions in the CREBBP Gene in Rubinstein–Taybi Syndrome Patients Using Multiplex PCR/Liquid Chromatography. Genet Test 10, 265-271CrossRefGoogle ScholarPubMed
65Kalkhoven, E. et al. (2003) Loss of CBP acetyltransferase activity by PHD finger mutations in Rubinstein–Taybi syndrome. Hum Mol Genet 12, 441-450CrossRefGoogle ScholarPubMed
66Udaka, T. et al. (2005) Comprehensive screening of CREB-binding protein gene mutations among patients with Rubinstein–Taybi syndrome using denaturing high-performance liquid chromatography. Congenit Anom 45, 125-131CrossRefGoogle ScholarPubMed
67Bartsch, O. et al. (2002) Molecular studies in 10 cases of Rubinstein–Taybi syndrome, including a mild variant showing a missense mutation in codon 1175 of CREBBP. J Med Genet 39, 496-501CrossRefGoogle ScholarPubMed
68Coupry, I. et al. (2002) Molecular analysis of the CBP gene in 60 patients with Rubinstein–Taybi syndrome. J Med Genet 39, 415-421CrossRefGoogle ScholarPubMed
69Murata, T. et al. (2001) Defect of histone acetyltransferase activity of the nuclear transcriptional coactivator CBP in Rubinstein–Taybi syndrome. Hum Mol Genet 10, 1071-1076CrossRefGoogle ScholarPubMed
70Zimmermann, N. et al. (2007) Confirmation of EP300 gene mutations as a rare cause of Rubinstein-Taybi syndrome. Eur J Hum Genet advance online publication 14 February 2007; doi: 10.1038/sj.ejhg.5201791Google ScholarPubMed
71Bito, H., Deisseroth, K. and Tsien, R.W. (1996) CREB phosphorylation and dephosphorylation: a Ca(2+)- and stimulus duration-dependent switch for hippocampal gene expression. Cell 87, 1203-1214CrossRefGoogle Scholar
72Abel, T. et al. (1997) Genetic demonstration of a role for PKA in the late phase of LTP and in hippocampus-based long-term memory. Cell 88, 615-626CrossRefGoogle ScholarPubMed
73Atkins, C.M. et al. (1998) The MAPK cascade is required for mammalian associative learning. Nat Neurosci 1, 602-609CrossRefGoogle ScholarPubMed
74Bourtchouladze, R. et al. (2003) A mouse model of Rubinstein–Taybi syndrome: defective long-term memory is ameliorated by inhibitors of phosphodiesterase 4. Proc Natl Acad Sci U S A 100, 10518-10522CrossRefGoogle ScholarPubMed
75Wood, M.A. et al. (2006) A transcription factor-binding domain of the coactivator CBP is essential for long-term memory and the expression of specific target genes. Learn Mem 13, 609-617CrossRefGoogle ScholarPubMed
76Oike, Y. et al. (1999) Mice homozygous for a truncated form of CREB-binding protein exhibit defects in hematopoiesis and vasculo-angiogenesis. Blood 93, 2771-2779CrossRefGoogle ScholarPubMed
77Alarcon, J.M. et al. (1999) Chromatin acetylation, memory, and LTP are impaired in CBP +/− mice: a model for the cognitive deficit in Rubinstein–Taybi syndrome and its amelioration. Neuron 42, 947-959CrossRefGoogle Scholar
78Korzus, E., Rosenfeld, M.G. and Mayford, M. (2004) CBP histone acetyltransferase activity is a critical component of memory consolidation. Neuron 42, 961-972CrossRefGoogle ScholarPubMed
79Wood, M.A. et al. (2005) Transgenic mice expressing a truncated form of CREB-binding protein (CBP) exhibit deficits in hippocampal synaptic plasticity and memory storage. Learn Mem 12, 111-119CrossRefGoogle ScholarPubMed
80Josselyn, S.A. (2005) What's right with my mouse model? New insights into the molecular and cellular basis of cognition from mouse models of Rubinstein–Taybi Syndrome. Learn Mem 12, 80-83CrossRefGoogle ScholarPubMed
81Gayther, S.A. et al. (2000) Mutations truncating the EP300 acetylase in human cancers. Nat Genet 24, 300-303CrossRefGoogle ScholarPubMed
82Ozdag, H. et al. (2002) Mutation analysis of CBP and PCAF reveals rare inactivating mutations in cancer cell lines but not in primary tumours. Br J Cancer 87, 1162-1165CrossRefGoogle ScholarPubMed
83Kishimoto, M. et al. (2005) Mutations and deletions of the CBP gene in human lung cancer. Clin Cancer Res 11, 512-519CrossRefGoogle ScholarPubMed
84Hennekam, R.C. (2006) Rubinstein–Taybi syndrome. Eur J Human Genet 14, 981-985CrossRefGoogle ScholarPubMed
85Wallerstein, R. et al. (1997) Submicroscopic deletions at 16p13.3 in Rubinstein–Taybi syndrome: frequency and clinical manifestations in a North American population. J Med Genet 34, 203-206CrossRefGoogle Scholar

Further reading, resources and contacts

The Online Mendelian Inheritance in Man (OMIM) website provides a description of Rubinstein–Taybi syndrome (Rubinstein–Taybi syndrome, OMIM 180849; CREBBP or CBP, OMIM 600140; EP300 or p300, OMIM 602700):

Gorlin, R.J., Cohen, M.M. and Hennekam, R.C.M. (2001). Syndromes of the Head and Neck. [Oxford Monographs on Medical Genetics (No. 42)], Oxford University Press, New YorkCrossRefGoogle Scholar
Petrij, F. et al. (2004) CBP and the Rubinstein–Taybi syndrome. In Inborn Errors of Development, 1st Edn (Epstein, CJ et al. , Eds), pp 728746, Oxford University Press, New YorkGoogle Scholar
Hallam, T.M. and Bourtchouladze, R. (2006) Rubinstein-Taybi syndrome: molecular findings and therapeutic approaches to improve cognitive dysfunction. Cell Mol Life Sci 63, 17251735.Google Scholar