doi:10.1086/302329 1216 Letters to the Editor Table 1 RET and EDNRB Mutations in 50 Male Patients with Isolated HSCR Patient Number Genea Exon Nucleotide Change Amino Acid Change Type of Case Length of Aganglionosis HSCR20 EDNRB 4 G1151A S305N Sporadic Short HSCR18 EDNRB 6 del1369A N378I Familial Short HSCR46 RET 10 C1876A Q626K Sporadic Ultrashort HSCR20 RET 11 C1941T I647I Sporadic Short HSCR25 RET 11 G1947A S649S Sporadic Short HSCR40 RET 14 G2438A R813Q Sporadic Short HSCR01 RET 14 G2607�5A Sporadic Long a For the RET, EDNRB, and EDN3 genes, primer sequences and PCR-SSCP conditions have been described elsewhere (Ceccherini et al. 1994; Auricchio et al. 1996). To screen the GDNF gene we used both SSCP (for exon 1) and denaturing gradient gel electrophoresis (DGGE) (for exon 2) with the following primer sets: 1F/R (5′-AGGCTTAACGTGCATTCTG-3′ and 5′- GGGAACGGTTCTTACAGT-3′), 2AF/R (5′-30-bp GC clamp-GATCATTTTTGTCTCATG- TGCCA-3′ and 5′-TCCTCTAATTCTCTGGGT-3′), 2BF/R (5′-GAGCGGAATCGGCAGGCTG-3′ and 5′-30-bp GC clamp-CAAGAGCCGCTGCAGTACCT-3′) and 2CF/R (5′-CTTGGGT- CTGGGCTATGAA-3′ and 5′-30-bp GC clamp-GCAATACACAGCAGTCTCTG-3′). PCR anneal- ing temperatures were 57�C–63�C. SSCP was done as reported elsewhere (Ceccherini et al. 1994), whereas a 20%–70% denaturing gradient in 1#TEA buffer at 160 V constant for 3.5–4.5 h was applied for DGGE analysis. Am. J. Hum. Genet. 64:1216–1221, 1999 Double Heterozygosity for a RET Substitution Interfering with Splicing and an EDNRB Missense Mutation in Hirschsprung Disease To the Editor: Hirschsprung disease (HSCR [MIM 142623]) is a de- velopmental disorder resulting from the arrest of the craniocaudal migration of enteric neurons from the neu- ral crest along gastrointestinal segments of variable length (Behrman 1992). The involvement of the Ret proto-oncogene and the endothelin-B receptor-mediated signaling pathways in the migration and differentiation of enteric ganglion cells has been demonstrated in both humans and mice. Heterozygous, incompletely penetrant point muta- tions and deletions of the RET proto-oncogene have been described in sporadic and familial cases of HSCR (Edery et al. 1994; Romeo et al. 1994; Angrist et al. 1995). In addition, homozygous RET-targeted disrup- tion in mice results in megacolon with renal abnormal- ities (Schuchardt et al. 1994). This phenotype is remi- niscent of the knockout for GDNF, encoding for the glial cell-line–derived neurotrophic growth factor (Moore et al. 1996; Pichel et al. 1996; Sanchez et al. 1996), a pro- tein that has been demonstrated to bind specifically and to activate Ret with a glycophosphatidylinositol (GPI)- anchored protein, the GDNF receptor-a (GFRA1) (Jing et al. 1996; Treanor et al. 1996). Although no nucleotide changes have been found at the GFRA1 locus (Angrist et al. 1998; Myers et al. 1998), rare heterozygous mu- tations of the GDNF gene, in some cases in combination with RET mutations, have been detected in a few pa- tients (Angrist et al. 1996; Ivanchuk et al. 1996; Salomon et al. 1996). Similarly, a missense mutation in the neur- turin (NTN) gene, a component of a second RET bind- ing complex including the NTN receptor (also known as GDNFR-a2, GDNFR-b, or GFRA2), has been found in an HSCR family cosegregating with a RET missense mutation (Doray et al. 1998). Rarely, megacolon pres- ents in association with pigmentary anomalies in the Shah-Waardenburg syndrome, a human autosomal re- cessive disease similar to the mouse piebald lethal and lethal spotting spontaneous mutants. The study of these models of syndromic megacolon has led to the identi- fication of homozygous mutations in the endothelin-B receptor gene (EDNRB) and the endothelin 3 (EDN3) gene in both humans and mice (Baynash et al. 1994; Hosoda et al. 1994; Puffenberger et al. 1994; Attié et al. 1995). In particular, in a large Mennonite pedigree, a founder homozygous W276C EDNRB mutation has been found in association with a specific RET haplotype, thus suggesting a possible genetic interaction between the two genes in the Mennonites affected with the Shah- Waardenburg syndrome (Puffenberger et al. 1994). EDNRB and EDN3, respectively, encode the endothelin- B receptor, a G-protein–coupled receptor with seven TM domains, and its ligand endothelin-3. We and others have demonstrated heterozygous incompletely penetrant mutations in the EDNRB gene in individuals with iso- lated megacolon (Amiel et al. 1996; Auricchio et al. 1996; Kusafuka et al. 1996), although mutations at the EDN3 locus are rarely found in HSCR (Bidaud et al. 1997). Recently, mutations of the transcriptional factor SOX10 (Pingault et al. 1998) and of the endothelin- Letters to the Editor 1217 Figure 1 SSCP analysis of the RET exon 11 transcript from affected individuals carrying the I647I silent mutation. PCR products corresponding to this exon from father, mother, and patient 1 genomic DNA are represented in lanes 1, 2, and 3, whereas lanes 4, 5, and 6 show the PCR products obtained from the cDNA of patients 1 and 2 and a control individual, respectively. Note that the shifted upper band corresponding to the mutated allele present in lanes 2 and 3 is absent in the cDNAs of patients 1 and 2 (lanes 4 and 5), which show only the normal allele present in the cDNA from a control individual (lane 6). Figure 2 ARMS analysis of the RET exon 11 from patient 2 genomic DNA and cDNA. PCR products were obtained by use of oligonucleotide primers specific for either the normal (lanes 1 and 3) or the I647I (lanes 2 and 4) RET alleles. A band of the expected size is amplified from patient genomic DNA with both sets of primers (lanes 1 and 2), whereas at the cDNA level the normal (lane 3), but not the mutated, allele (lane 4) is present. converting enzyme 1 (ECE1) (Hofstra et al. 1998) have been reported in HSCR patients with pigmentary and cardiac defects. RET, GDNF, EDNRB, and EDN3 are mutated in a variable proportion of individuals affected with isolated HSCR, accounting for 30%–50% of all cases (Edery et al. 1997; Eng and Mulligan 1997; Hofstra et al. 1997). In addition, these mutations alone do not explain the variable expressivity and the incomplete penetrance of the disease for which, according to a complex model of inheritance, the combined presence of mutations in more than one of the known or still unknown HSCR suscep- tibility genes can be inferred. To better understand the contribution of RET, GDNF, EDNRB, and EDN3 to the pathogenesis of HSCR, we collected samples from 50 patients and analyzed them for mutations at these loci. Forty-seven of these patients represented unrelated sporadic cases, and three belonged to two different families in which a polygenic or recessive mode of inheritance could be inferred on the basis of the pedigree. They were affected with isolated mega- colon, including long, short (“classic”), and ultrashort forms. None of them was born of consanguineous par- ents, and all were of Italian origin. Table 1 shows two heterozygous incompletely pene- trant EDNRB mutations (S305N and N378I) we re- cently described in two patients with isolated short-seg- ment HSCR disease, inherited from healthy parents (Auricchio et al. 1996). These two mutations were iden- tified during a first screening of 20 patients and were absent in 100 control chromosomes. We then analyzed samples from 30 additional patients with isolated HSCR and could not detect any novel mutations at the EDNRB locus. The mutation screening of the 21 exons of the RET proto-oncogene was done on all 50 patients. As shown in table 1, two missense mutations in exons 10 and 14 (C1876A and G2438A, respectively) and a nu- cleotide change affecting the 5′ end of intron 14 (G2607�5A) were identified in three different patients. In particular, the Q626K missense mutation affects the last codon of exon 10, namely the cys-rich region located in the juxtamembranous extracellular domain, suggest- ing either an effect on the folding of the ligand-binding domain or an interference with RET dimerization, a cru- cial step preceding its activation. Mutation R813Q, af- fecting exon 14, is likely to alter the RET tyrosine kinase activity, whereas the GrA transition at the 5′ end of intron 14 might interfere with correct processing of the primary transcript. In addition, we found two synony- mous nucleotide changes in RET exon 11, leading to neutral substitutions I647I and S649S in patients 20 and 25, respectively (table 1). These were absent in 150 con- trol individuals. We could not detect any GDNF or EDN3 mutations in our patients, thus confirming that they play a minor role in HSCR pathogenesis. 1218 Letters to the Editor Figure 3 In vitro analysis of the RET exons 10 and 11 splicing products from both the normal and the I647I mutated alleles (see text for details). a, Schematic representation of the RET constructs used for the in vitro splicing analysis. b, RT-PCR amplification using primers designed in exons 10 and 11, as indicated in a, from total RNA of COS-7 cells transiently transfected with the RET wild-type (lane 1) and the I647I (lane 2) constructs. Note the abnormal products present in lane 2. c, Sequence analysis of the 255-bp products from lanes 1 and 2 shown in b corresponded to the normal and mutated RET alleles, respectively, an observation consistent with the use of canonical “gt” and “ag” donor and acceptor splice sites (underlined in the partial intronic sequence given in lowercase letters). Conversely, sequence analysis of the 148- bp and 120-bp products revealed two exon 11 deleted forms resulting from the use of cryptic acceptor “ag” sites activated to generate the two abnormal products (underlined in the sequence of the 5′ portion of exon 11 given in capital letters). The two alternative nucleotides involved in the neutral substitution of codon 647 (T in the normal allele and C in the variant allele) are shown in boldface type. Interestingly, patient 20 (from now on, patient 1) car- ries both a RET synonymous nucleotide change, C1941T (I647I), inherited from the unaffected mother, and an EDNRB missense mutation, S305N, transmitted by the healthy father. This latter mutation results in the substitution of a serine, located in the third intracellular loop, with an asparagine (Auricchio et al. 1996). This serine, highly conserved between different species, has been shown to be a site of in vivo phosphorylation (Roos et al. 1998), suggesting that it plays an important role in the receptor regulation and that its absence can result in the protein loss of function. Intraexonic silent mu- tations can alter correct mRNA processing, thus result- ing in either altered mRNA levels or truncated proteins (Steingrimsdottir et al. 1992; De Meirleir et al. 1994; Li et al. 1995; Richard and Beckmann 1995; Jin et al. 1996; Llewellyn et al. 1996; Ploos van Amstel et al. 1996; Liu et al. 1997). To test the hypothesis that the RET I647I variant could play a causative role in the development of the disease phenotype in patient 1, we analyzed, both in vitro and in vivo, the RET mRNA expression, using cDNA and genomic DNA from patient 1 and from an additional proband (patient 2) with the same I647I change, described elsewhere (Ceccherini et al. 1994). As an in vivo approach to study the RET transcrip- tion, we performed reverse transcription (RT) PCR with total RNA from lymphoblastoid cell lines of these two patients, by means of two different techniques—the SSCP (Orita et al. 1989) and the amplification-refractory mutation system (ARMS) (Newton et al. 1989) analysis. Results obtained from SSCP analysis of the RET exon 11 are shown in figure 1. The shifted upper band cor- responding to the mutated allele present in the genomic DNA of patient 1 and his healthy mother (lanes 2 and 3) is absent in the cDNAs of patients 1 and 2 (lanes 4 and 5). They depict only the normal allele present in the cDNA from a control individual (lane 6). Similar results have been obtained from ARMS analysis of the RET exon 11 from patient 2 genomic DNA and cDNA (fig. 2). In particular, using oligonucleotide primers specific for either the normal (lanes 1 and 3) or the I647I (lanes 2 and 4) RET alleles, we were able to amplify a band Letters to the Editor 1219 of the expected size from patient genomic DNA with both sets of primers (lanes 1 and 2), although at the cDNA level the normal (lane 3), but not the mutated, allele (lane 4) was present. In addition, direct sequence analysis of the PCR product, as well as BsmI enzymatic restriction of an exon 7 RET polymorphism, the phase of which is known with respect to the I647I mutation, confirmed the absence of the mutated allele in the cDNA of the affected individuals (data not shown). To elucidate whether the I647I silent mutation could interfere with splicing, thus resulting in unstable prod- ucts undetectable in vivo in lymphoblast cDNAs, we cloned the 3-kb genomic region encompassing RET ex- ons 10, 11, and 12, from both the normal and the I647I mutated alleles, into the pSPL3 eukaryotic vector (Church et al. 1994) (fig. 3a). RT-PCR amplification from total RNA of SV40-transformed African green monkey kidney cells (COS-7) transiently transfected with the RET wild-type construct (fig. 3b) shows the expected fragment of 255 bp (lane 1). Two additional products of 148 bp and 120 bp were amplified when the I647I construct was transfected (lane 2). As shown in figure 3c, sequence analysis of the 255-bp products from the above lanes 1 and 2 corresponded to the normal and mutated RET alleles, respectively, an observation consistent with the use of canonical “gt” and “ag” donor and acceptor splice sites (underlined in the partial in- tronic sequence given in lowercase letters). Conversely, sequence analysis of the 148-bp and 120-bp products revealed two exon 11 deleted forms. Both products re- sulted from the fusion of the 3′ end of exon 10 to two different sites located 107 bp and 135 bp downstream of the exon 11 starting nucleotide in the largest and the smallest abnormal products, respectively. If translated, these mRNAs would result in two proteins: the one cor- responding to the largest transcript truncated 758 bp downstream of the newly activated splice site, and the smallest mRNA resulting in an in-frame interstitial loss of 45 amino acids, including the transmembrane do- main. Nevertheless, we postulate that this abnormal pro- cessing of the I647I allele may reduce the mutant RNA to a level that escapes detection by the commonly used in vivo RT-PCR approach. Thus, the presence in our patient of an inactive form of the endothelin-B receptor and of reduced levels of the Ret protein could impair the normal enteric neuronal migration. In conclusion, we confirm that in isolated HSCR the major susceptibility locus is the RET proto-oncogene, with EDNRB accounting for a minority of cases. More relevantly, we demonstrate in two different patients, both in vivo and in vitro, that the same silent RET mu- tation can interfere with correct transcription, thus pos- sibly leading to a reduced level of the Ret protein. Finally, the coexistence, reported for the first time, in the same patient of two functionally significant EDNRB and RET mutations suggests a direct genetic interaction between these two distinct transmembrane receptors in polygenic HSCR disease. Acknowledgments The financial support of Telethon–Italy (grant E791) is grate- fully acknowledged. This work was also funded by the Italian Telethon Foundation, the Italian Ministry of Health, and the European Community (contract MH4-CT97-2107). ALBERTO AURICCHIO,1,∗ PAOLA GRISERI,2 MARIA LUISA CARPENTIERI,3 NICOLA BETSOS,2 ANNAMARIA STAIANO,3 ARTURO TOZZI,3 MANUELA PRIOLO,2 HELEN THOMPSON,2 RENATA BOCCIARDI,2 GIOVANNI ROMEO,4,5 ANDREA BALLABIO,1 AND ISABELLA CECCHERINI2 1Telethon Institute of Genetics and Medicine, Milan, 2Laboratorio di Genetica Molecolare, Istituto Giannina Gaslini, Genova-Quarto, 3Dipartimento di Pediatria, Università �Federico II,� Naples, and 4Dipartimento di Oncologia Biologia e Genetica, Università degli Studi di Genova, Genova, Italy; and 5Genetic Cancer Susceptibility Unit, Lyon, France Electronic-Database Information Accession number and URL for data in this study are as follows: Online Mendelian Inheritance in Man (OMIM), http:// www.ncbi.nlm.nih.gov/Omim (for HSCR [142623]) References Amiel J, Attié T, Jan D, Pelet A, Edery P, Bidaud C, Lacombe D, et al (1996) Heterozygous endothelin receptor B (EDNRB) mutations in isolated Hirschsprung disease. Hum Mol Genet 5:355–357 Angrist M, Bolk S, Halushka M, Lapchak PA, Chakravarti A (1996) Germline mutations in glial cell line-derived neuro- trophic factor (GDNF) and RET in a Hirschsprung disease patient. Nat Genet 14:341–344 Angrist M, Bolk S, Thiel B, Puffenberger EG, Hofstra RM, Buys CHCM, Cass DT, et al (1995) Mutation analysis of the RET receptor tyrosine kinase in Hirschsprung disease. Hum Mol Genet 4:821–830 Angrist M, Jing S, Bolk S, Bentley K, Nallasamy S, Halushka M, Fox GM, et al (1998) Human GFRA1: cloning, mapping, genomic structure, and evaluation as a candidate gene for Hirschsprung disease susceptibility. Genomics 48:354–362 Attié T, Till M, Pelet A, Amiel J, Edery P, Boutrand L, Munnich A, et al (1995) Mutations of the endothelin-receptor B gene in Waardenburg-Hirschsprung disease. Hum Mol Genet 4: 2407–2409. Auricchio A, Casari G, Staiano A, Ballabio A (1996) Endothe- lin-B receptor mutations in patients with isolated Hirsch- 1220 Letters to the Editor sprung disease from a non-inbred population. Hum Mol Genet 5:351–354 Baynash AG, Hosoda K, Giaid A, Richardson JA, Emoto N, Hammer RE, Yanagisawa M (1994) Interaction of endo- thelin-3 with endothelin-B receptor is essential for devel- opment of epidermal melanocytes and enteric neurons. Cell 79:1277–1285 Behrman RE (1992) Nelson textbook of pediatrics, 14th ed. W. B. Saunders, Philadelphia, pp 954–956 Bidaud C, Salomon R, Van Camp G, Pelet A, Attie T, Eng C, Bonduelle M, et al (1997) Endothelin-3 gene mutations in isolated and syndromic Hirschsprung disease. Eur J Hum Genet 5:247–251 Ceccherini I, Hofstra RM, Luo Y, Stulp RP, Barone V, Stel- wagen T, Bocciardi R, et al (1994) DNA polymorphisms and conditions for SSCP analysis of the 20 exons of the ret proto-oncogene. Oncogene 9:3025–3029 Church DM, Stotler CJ, Rutter JL, Murrell JR, Trofatter JA, Buckler AJ (1994) Isolation of genes from complex sources of mammalian genomic DNA using exon amplification. Nat Genet 6:98–105 De Meirleir L, Lissens W, Benelli C, Ponsot G, Desguerre I, Marsac C, Rodriguez D, et al (1994) Aberrant splicing of exon 6 in the pyruvate dehydrogenase-E1 alpha mRNA linked to a silent mutation in a large family with Leigh’s encephalomyelopathy. Pediatr Res 36:707–712 Doray B, Salomon R, Amiel J, Pelet A, Touraine R, Billaud M, Attie T, et al (1998) Mutation of the RET ligand, neur- turin, supports multigenic inheritance in Hirschsprung dis- ease. Hum Mol Genet 7:1449–1452 Edery P, Eng C, Munnich A, Lyonnet S (1997) RET in human development and oncogenesis. BioEssays 19:389–395 Edery P, Lyonnet S, Mulligan LM, Pelet A, Dow E, Abel L, Holder S, et al (1994) Mutations of the RET proto-oncogene in Hirschsprung’s disease. Nature 367:378–379 Eng C, Mulligan LM (1997) Mutations of the RET proto- oncogene in the multiple endocrine neoplasia type 2 syn- dromes, related sporadic tumours, and Hirschsprung dis- ease. Hum Mutat 9:97–109 Hofstra R, Osinga J, Buys C (1997) Mutations in Hirschsprung disease: when does a mutation contribute to the phenotype. Eur J Hum Genet 5:180–185 Hofstra R, Valdenaire O, Arch E, Osinga J, Meijers C, Buys CHCM (1998) A loss of function mutation in the endo- thelin-converting enzyme 1 in a patient with Hirschsprung disease and cardiac defects. Paper presented at the Third International Meeting on Hirschsprung Disease and Related Neurocristopathies. Evian, France, February 5–8 Hosoda K, Hammer RE, Richardson JA, Baynash AG, Cheung JC, Giaid A, Yanagisawa M (1994) Targeted and natural (piebald-lethal) mutations of endothelin-B receptor gene produce megacolon associated with spotted coat colon in mice. Cell 79:1267–1276 Ivanchuk SM, Myers SM, Eng C, Mulligan LM (1996) De novo mutation of GDNF, ligand for the RET/GDNFR-alpha receptor complex, in Hirschsprung disease. Hum Mol Genet 5:2023–2026 Jin Y, Dietz HC, Montgomery RA, Bell WR, McIntosh I, Coller B, Bray PF (1996) Glanzmann thrombasthenia: cooperation between sequence variants in cis during splice site selection. J Clin Invest 98:1745–1754 Jing S, Wen D, Yu Y, Holst PL, Luo Y, Fang M, Tamir R, et al (1996) GDNF-induced activation of the ret protein ty- rosine kinase is mediated by GDNFR-alpha, a novel receptor for GDNF. Cell 85:1113–1124 Kusafuka T, Wang Y, Puri P (1996) Novel mutations of the endothelin-B receptor gene in isolated patients with Hirschs- prung’s disease. Hum Mol Genet 5:347–349 Li X, Park W-J, Pyertz RE, Jabs WE (1995) Effect on splicing of a silent FGFR2 mutation in Crouzon syndrome. Nat Ge- net 9:232–233 Liu W, Qian C, Franke U (1997) Silent mutation induces exon skipping of fibrillin-1 gene in Marfan syndrome. Nat Genet 16:328–329 Llewellyn DH, Scobie GA, Urquhart AJ, Whatley SD, Roberts AG, Harrison PR, Elder GH (1996) Acute intermittent por- phyria caused by defective splicing of porphobilinogen de- aminase RNA: a synonymous codon mutation at �22 bp from the 5′ splice site causes skipping of exon 3. J Med Genet 33:437–438 Moore MW, Klein RD, Farinas I, Sauer H, Armanini M, Phil- lips H, Reichard LF, et al (1996) Renal and neuronal ab- normalities in mice lacking GDNF. Nature 382:76–79 Myers SM, Salomon R, Goessling A, Pelet A, von Deimling A, Lyonnet S, Mulligan LM (1998) Analysis of the GFRA family in Hirschsprung disease. Paper presented at the Third International Meeting on Hirschsprung Disease and Related Neurocristopathies. Evian, France, February 5–8 Newton CR, Graham A, Heptinstall LE, Powell SJ, Summers C, Kalsheker N, Smith JC, et al (1989) Analysis of any point mutation in DNA: the amplification refractory mutation sys- tem (ARMS). Nucleic Acids Res 17:2503–2516 Orita M, Suzuki Y, Sekiya T, Hayashi K, (1989) Rapid and sensitive detection of point mutations and DNA polymor- phisms using the polymerase chain reaction. Genomics 5: 874–879 Pichel JG, Shen L, Sheng HZ, Granholm AC, Drago J, Grin- berg A, Lee EJ, et al (1996) Defects in enteric innervation and kidney development in mice lacking GDNF. Nature 382: 73–76 Pingault V, Bondurand N, Kuhlbrodt K, Goerich DE, Prehu MO, Puliti A, Herbarth B, et al (1998) SOX10 mutations in patients with Waardenburg-Hirschsprung disease. Nat Genet 18:171–173 Ploos van Amstel JK, Bergman AJ, van Beurden EA, Roijers JF, Peelen T, van den Berg IE, Poll-The BT, et al (1996) Hereditary tyrosinemia type 1: novel missense, nonsense and splice consensus mutations in the human fumarylacetoace- tate hydrolase gene; variability of the genotype-phenotype relationship. Hum Genet 97:51–59 Puffenberger EG, Hosoda K, Washington SS, Nakao K, de Wit D, Yanagisawa M, Chakravarti A (1994) A missense mu- tation of the endothelin-B receptor gene in multigenic Hirschsprung disease. Cell 79:1257–1266 Richard I, Beckmann JS, (1995) How neutral are synonymous codon mutations? Nat Genet 10:259 Romeo G, Ronchetto P, Yin L, Barone V, Seri M, Ceccherini I, Pasini B, et al (1994) Point mutations affecting the tyrosine Letters to the Editor 1221 kinase domain of the RET proto-oncogene in Hirschsprung’s disease. Nature 367:377–378 Roos M, Soskic V, Poznanovic S, Godovac-Zimmermann J (1998) Post-translational modifications of endothelin recep- tor B from bovine lungs analyzed by mass spectrometry. J Biol Chem 273:924–931 Salomon R, Attié T, Pelet A, Bidaud C, Eng C, Amiel J, Sar- nacki S, et al (1996) Germline mutations of the RET ligand GDNF are not sufficient to cause Hirschsprung disease. Nat Genet 14:345–347 Sanchez MP, Silos-Santiago I, Frisen J, He B, Lira SA, Barbacid M (1996) Renal agenesis and the absence of enteric neurons in mice lacking GDNF. Nature 382:70–73 Schuchardt A, D’Agati V, Larsson-Blomberg L, Costantini F, Pachnis V (1994) Defects in the kidney and enteric nervous system of mice lacking the tyrosine kinase receptor Ret. Na- ture 367:380– 383 Steingrimsdottir H, Rowley G, Dorado G, Cole J, Lehmann AR (1992) Mutations which alter splicing in the human hypoxanthine-guanine phosphoribosyltransferase gene. Nu- cleic Acids Res 20:1201–1208 Treanor JJ, Goodman L, de Sauvage F, Stone DM, Poulsen KT, Beck CD, Gray C, et al (1996) A characterization of a multicomponent receptor for GDNF. Nature 382:80–83 Address for correspondence and reprints: Dr. Isabella Ceccherini, Laboratorio di Genetica Molecolare, Istituto Giannina Gaslini, Largo G. Gaslini, 5, 16148 Genova, Italy. E-mail: isa.c@unige.it � 1999 by The American Society of Human Genetics. All rights reserved. 0002-9297/99/6404-0035$02.00 Am. J. Hum. Genet. 64:1221–1225, 1999 A Mutation (2314delG) in the Usher Syndrome Type IIA Gene: High Prevalence and Phenotypic Variation To the Editor: Usher syndrome (USH), an autosomal recessive condi- tion, is characterized by hearing impairment associated with retinitis pigmentosa (RP). It is a clinically and ge- netically heterogeneous condition. Three clinical forms of USH have been described, and eight loci have been mapped (Hereditary Hearing Loss home page). USH type I (USH1) is the most severe form, manifested by profound congenital deafness, constant vestibular dys- function, and prepubertal onset of RP. USH type II (USH2) is characterized by congenital moderate-to-se- vere hearing impairment, normal vestibular responses, and RP (Smith et al. 1994). USH type III (USH3) is characterized by progressive hearing loss, variable ves- tibular problems, and RP (Pakarinen et al. 1995). The USH1B gene has been identified (Weil et al. 1995), and a variety of mutations leading to USH1 have been cat- alogued in the MYO7A gene (Liu et al. 1998). USH2 is the most common of the three types of USH, accounting for more than half of all cases (Hopes et al. 1997; Ro- senberg et al. 1997). Recently, Eudy et al. (1998) reported the identification of the USH type IIA gene (USH2A; MIM 276901). USH2A encodes a putative extracellular matrix protein of 1,551 amino acids with laminin epidermal growth factor and fibronectin type II motifs. Expression of the USH2A gene has been detected by reverse transcriptase PCR (RT-PCR) from fetal human cochlea, eye, and adult human retina (Eudy et al. 1998). Three different mu- tations were identified in patients with USH2A, the most frequent of which is the 2314delG mutation. Of 96 pro- bands tested, 21 carried the 2314delG mutation. In the present study, we have undertaken mutational analysis of the USH2A gene in 23 families with USH (both USH2 and atypical USH), from the United King- dom and China. We found that the majority of families with USH2 carry the 2314delG mutation. Surprisingly, we determined that the 2314delG mutation in the USH2A gene can also lead to atypical USH. Of 23 families with USH analyzed in this study, 15 have been described elsewhere (Hopes et al. 1997). All available affected subjects were examined by one of the authors. Full clinical histories were obtained, with em- phasis on any potential causes of hearing impairment. The audiovestibular and ophthalmic evaluations were done on the basis of recommendations by the Usher Syndrome Consortium (Smith et al. 1994). The details of the clinical evaluations can be found in Hopes et al. (1997). The cases studied here are classified clinically into the following two groups: (1) USH2, consisting of congenital sloping moderate/severe hearing impairment, normal vestibular function, and RP (Smith et al. 1994); and (2) atypical USH, consisting of bilateral sensorineu- ral progressive hearing loss without other obvious fac- tors as the cause of the progression of the hearing im- pairment, along with variable vestibular dysfunction and RP. Of 23 families studied, 13 were given a diagnosis of USH2 and 10 were given a diagnosis of atypical USH. Eighteen families living in the United Kingdom and five in China were included in our analyses. Twenty-three probands with USH were screened for the 2314delG mutation by combined SSCP/heteroduplex analysis. Twelve families (52%) carried the 2314delG mutation— 8 (62%) of 13 patients with typical USH2 and 4 (40%) of 10 patients with atypical USH (table 1). Of 12 families with this mutation, 7 were identified with only one af- fected member (sporadic cases). Of the remaining five families with more than one affected individual, two families (USH.05 and USH.10) with two affected sibs were tested for cosegregation of the disease with the USH2A locus (Eudy et al. 1998). The segregation anal- ysis was consistent with linkage to USH2A (data not Double Heterozygosity for a RET Substitution Interfering with Splicing and an EDNRB Missense Mutation in Hirschsprung Disease Acknowledgments References