Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Genes (Basel) ; 15(5)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38790165

ABSTRACT

Adams-Oliver syndrome is a rare inherited condition characterized by scalp defects and limb abnormalities. It is caused by variants in different genes such as ARHGAP31. Here, we used an interdisciplinary approach to study a family with lower limb anomalies. We identified a novel variant in the ARHGAP31 gene that is predicted to result in a truncated protein with a constitutively activated catalytic site due to the loss of 688 amino acids involved in the C-terminal domain, essential for protein auto-inhibition. Pathogenic variants in ARHGAP31 exon 12, leading to a premature protein termination, are associated with Adams-Oliver syndrome. Bioinformatic analysis was useful to elucidate the impact of the identified genetic variant on protein structure. To better understand the impact of the identified variant, 3D protein models were predicted for the ARHGAP31 wild type, the newly discovered variant, and other pathogenetic alterations already reported. Our study identified a novel variant probably involved in Adams-Oliver syndrome and increased the evidence on the phenotypic variability in patients affected by this syndrome, underlining the importance of translational research, including experimental and bioinformatics analyses. This strategy represents a successful model to investigate molecular mechanisms involved in syndrome occurrence.


Subject(s)
Ectodermal Dysplasia , GTPase-Activating Proteins , Pedigree , Phenotype , Scalp Dermatoses , Humans , GTPase-Activating Proteins/genetics , Scalp Dermatoses/genetics , Scalp Dermatoses/congenital , Scalp Dermatoses/pathology , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/pathology , Male , Female , Mutation , Limb Deformities, Congenital/genetics , Phosphoproteins
3.
Eur J Pediatr ; 183(5): 2257-2272, 2024 May.
Article in English | MEDLINE | ID: mdl-38411716

ABSTRACT

Patients with Aarskog-Scott syndrome (AAS) have short stature, facial anomalies, skeletal deformities, and genitourinary malformations. FYVE, RhoGEF, and PH domain-containing 1 (FGD1) is the only known causative gene of AAS. However, the diagnosis of AAS remains difficult, and specific treatments are still absent. Patients suspected with AAS were recruited, and clinical information was collected. Genetic testing and functional analysis were carried out for the diagnosis. By literature review, we summarized the clinical and genetic characteristics of FGD1-related AAS and analyzed the genotype-phenotype correlation. Five patients were recruited, and four novel FGD1 variants were identified. The diagnosis of AAS was confirmed by genetic analysis and functional study. Three patients treated with growth hormone showed improved heights during the follow-up period. By literature review, clinical features of AAS patients with FGD1 variants were summarized. Regarding FGD1 variations, substitutions were the most common form, and among them, missense variants were the most frequent. Moreover, we found patients with drastic variants showed higher incidences of foot and genitourinary malformations. Missense variants in DH domain were related to a lower incidence of cryptorchidism.   Conclusion: We reported four novel pathogenic FGD1 variations in AAS patients and confirmed the efficacy and safety of growth hormone treatment in FGD1-related AAS patients with growth hormone deficiency. Additionally, our literature review suggested the crucial role of DH domain in FGD1 function. What is Known: • Aarskog-Scott syndrome is a rare genetic disease, and the only known cause is the variant in FGD1 gene. The typical clinical manifestations of AAS include facial, skeletal, and urogenital deformities and short stature. What is New: • We reported four novel FGD1 variants and reported the treatment of growth hormone in FGD1-related AAS patients. Our genotype-phenotype correlation analysis suggested the crucial role of DH domain in FGD1 function.


Subject(s)
Abnormalities, Multiple , Face/abnormalities , Genetic Diseases, X-Linked , Genitalia, Male/abnormalities , Guanine Nucleotide Exchange Factors , Humans , Guanine Nucleotide Exchange Factors/genetics , Male , Female , Child, Preschool , Abnormalities, Multiple/genetics , Abnormalities, Multiple/diagnosis , Child , Infant , Heart Defects, Congenital/genetics , Heart Defects, Congenital/diagnosis , Urogenital Abnormalities/genetics , Urogenital Abnormalities/diagnosis , Genetic Association Studies , Dwarfism/genetics , Dwarfism/diagnosis , Dwarfism/drug therapy , Scalp Dermatoses/genetics , Scalp Dermatoses/diagnosis , Scalp Dermatoses/drug therapy , Scalp Dermatoses/congenital , Phenotype , Hand Deformities, Congenital/genetics , Hand Deformities, Congenital/diagnosis
4.
Am J Med Genet A ; 191(6): 1593-1598, 2023 06.
Article in English | MEDLINE | ID: mdl-36866832

ABSTRACT

The Notch proteins play key roles in cell fate determination during development. Germline pathogenic variants in NOTCH1 predispose to a spectrum of cardiovascular malformations including Adams-Oliver syndrome and a wide variety of isolated complex and simple congenital heart defects. The intracellular C-terminus of the single-pass transmembrane receptor encoded by NOTCH1 contains a transcriptional activating domain (TAD) required for target gene activation and a PEST domain (a sequence rich in proline, glutamic acid, serine, and threonine), regulating protein stability and turnover. We present a patient with a novel variant encoding a truncated NOTCH1 protein without the TAD and PEST domain (NM_017617.4: c.[6626_6629del];[=], p.(Tyr2209CysfsTer38)) and extensive cardiovascular abnormalities consistent with a NOTCH1-mediated mechanism. This variant fails to promote transcription of target genes as assessed by luciferase reporter assay. Given the roles of the TAD and PEST domains in NOTCH1 function and regulation, we hypothesize that loss of both the TAD and the PEST domain results in a stable, loss-of-function protein that acts as an antimorph through competition with wild-type NOTCH1.


Subject(s)
Ectodermal Dysplasia , Limb Deformities, Congenital , Scalp Dermatoses , Humans , Receptor, Notch1/genetics , Ectodermal Dysplasia/genetics , Scalp Dermatoses/congenital , Limb Deformities, Congenital/genetics
5.
Pediatr Dermatol ; 40(1): 212-213, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36178276

ABSTRACT

Cutis verticis gyrata (CVG), characterized by cerebriform overgrowth of the scalp, is rarely observed in congenital melanocytic nevi (CMN). We describe a 13-year-old male with autism and a large CMN of the scalp with numerous satellite nevi whose scalp nevus exhibited evolution with poliosis and CVG. Given the potential association of CVG (independent of CMN) with seizures, neuropsychiatric, and ophthalmologic disorders, and nevus-associated CVG (cerebriform intradermal nevus) with melanoma, multidisciplinary evaluation of CMN patients with CVG is important to guide management and treatment.


Subject(s)
Hair Diseases , Nevus, Pigmented , Pigmentation Disorders , Scalp Dermatoses , Skin Neoplasms , Male , Humans , Adolescent , Skin Neoplasms/complications , Skin Neoplasms/congenital , Scalp , Scalp Dermatoses/complications , Scalp Dermatoses/congenital , Nevus, Pigmented/complications
6.
Am J Med Genet A ; 188(11): 3318-3323, 2022 11.
Article in English | MEDLINE | ID: mdl-36059114

ABSTRACT

Aplasia cutis congenita (ACC) of the scalp and terminal transverse limb defects (TTLD) are the characteristic findings of Adams-Oliver syndrome (AOS). The variable clinical spectrum further includes cardiac, neurologic, renal, and ophthalmological findings. Associated genes in AOS are in the Notch and the CDC42/Rac1 signaling pathways. Both autosomal-dominant and autosomal-recessive inheritances have been reported, the latter with pathogenic variants in DOCK6 or EOGT. The EOGT-associated recessive type of AOS has been postulated to present a more favorable prognosis. We here report a 12-year-old girl from a refugee family of Iraq with consanguineous parents. She was born with a severe phenotype of AOS presenting a large ACC of the scalp with an underlying skull defect, which was often infected and inflamed. Afterward, additional ulceration developed. Furthermore, the girl showed microcephaly, TTLD on both hands and feet, and neurological findings: spastic paresis, epilepsy and suspicion of intellectual deficit. Molecular genetic analysis (next-generation sequencing) revealed a novel frameshift mutation in the EOGT gene in Exon 13 in homozygous constellation: c.1013dupA p.(Asn338Lysfs*24). A biopsy within an ulceration at the scalp ACC showed a cutaneous squamous cell carcinoma (cSCC) with local invasive growth into the dura, the meninges, and the cortex. Treatment including surgical resection and focal irradiation was not curative and the girl deceased 6 months after initial diagnosis. This report on a patient with AOS and an autosomal-recessive EOGT gene variant dying of a local aggressive cSCC at an ACC lesion shows that close monitoring of ACC is essential.


Subject(s)
Carcinoma, Squamous Cell , Ectodermal Dysplasia , Limb Deformities, Congenital , Scalp Dermatoses , Skin Neoplasms , Ectodermal Dysplasia/diagnosis , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/pathology , Female , Frameshift Mutation , Humans , Limb Deformities, Congenital/genetics , Mutation , N-Acetylglucosaminyltransferases/genetics , Scalp/pathology , Scalp Dermatoses/congenital , Scalp Dermatoses/diagnosis , Scalp Dermatoses/genetics , Scalp Dermatoses/pathology , Skin Neoplasms/diagnosis , Skin Neoplasms/genetics , Skull/pathology
7.
PLoS Genet ; 18(8): e1010335, 2022 08.
Article in English | MEDLINE | ID: mdl-35951645

ABSTRACT

Notch signaling is a conserved pathway that converts extracellular receptor-ligand interactions into changes in gene expression via a single transcription factor (CBF1/RBPJ in mammals; Su(H) in Drosophila). In humans, RBPJ variants have been linked to Adams-Oliver syndrome (AOS), a rare autosomal dominant disorder characterized by scalp, cranium, and limb defects. Here, we found that a previously described Drosophila Su(H) allele encodes a missense mutation that alters an analogous residue found in an AOS-associated RBPJ variant. Importantly, genetic studies support a model that heterozygous Drosophila with the AOS-like Su(H) allele behave in an opposing manner to heterozygous flies with a Su(H) null allele, due to a dominant activity of sequestering either the Notch co-activator or the antagonistic Hairless co-repressor. Consistent with this model, AOS-like Su(H) and Rbpj variants have decreased DNA binding activity compared to wild type proteins, but these variants do not significantly alter protein binding to the Notch co-activator or the fly and mammalian co-repressors, respectively. Taken together, these data suggest a cofactor sequestration mechanism underlies AOS phenotypes associated with RBPJ variants, whereby the AOS-associated RBPJ allele encodes a protein with compromised DNA binding activity that retains cofactor binding, resulting in Notch target gene dysregulation.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Co-Repressor Proteins , DNA , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/metabolism , Ectodermal Dysplasia , Humans , Limb Deformities, Congenital , Mammals/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Scalp/metabolism , Scalp Dermatoses/congenital , Skull/metabolism
8.
Am J Med Genet A ; 188(2): 658-664, 2022 02.
Article in English | MEDLINE | ID: mdl-34755929

ABSTRACT

Adams-Oliver syndrome (AOS), a rare inherited disorder, is characterized by scalp and terminal limb defects. Several genes associated with Notch pathway mutations have led to AOS. Here, we report a Thai male newborn presenting with aplasia cutis congenita and absence of a right pulmonary artery, which is suggestive of AOS. This was confirmed by the identification of a novel missense mutation in DLL4, a heterozygous one base pair change at nucleotide 82 (c.82G>C, p.Gly28Arg), which is in N-terminal domain. This is the first DLL4-related AOS case with arterial defect.


Subject(s)
Ectodermal Dysplasia , Limb Deformities, Congenital , Scalp Dermatoses , Adaptor Proteins, Signal Transducing/genetics , Calcium-Binding Proteins/genetics , Ectodermal Dysplasia/diagnosis , Ectodermal Dysplasia/genetics , Humans , Infant, Newborn , Limb Deformities, Congenital/diagnosis , Limb Deformities, Congenital/genetics , Male , Mutation , Pulmonary Artery , Scalp , Scalp Dermatoses/congenital , Scalp Dermatoses/diagnosis , Scalp Dermatoses/genetics
9.
Stem Cells Dev ; 30(12): 611-621, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33899511

ABSTRACT

Heterozygous loss-of-function mutation in Delta-like ligand-4 (Dll4) is an important cause of Adams-Oliver syndrome (AOS). Cardiac defects, in particular outflow tract (OFT) alignment defects, are observed in about one-fourth of patients with this syndrome. The mechanism underlying this genotype-phenotype correlation has not yet been established. Dll4-mediated Notch signaling is known to play a crucial role in second heart field (SHF) progenitor cell proliferation. We hypothesized that the depletion of the SHF progenitor pool of cells due to partial loss of Dll4 is responsible for the OFT alignment defects seen in AOS. To demonstrate this, we studied Dll4 expression by murine SHF progenitor cells around E9.5, a crucial time-point in SHF biology. We used SHF-specific (Islet1-Cre) conditional knockout of Dll4 to bypass the early embryonic lethality seen in global Dll4 heterozygotes. Dll4-mediated Notch signaling is critically required for SHF proliferation such that Dll4 knockout results in a 33% reduction in proliferation and a fourfold increase in apoptosis in SHF cells, leading to a 56% decline in the size of the SHF progenitor pool. A reduction in SHF cells available for incorporation into the developing heart leads to underdevelopment of the SHF-derived right ventricle and OFT. Similar to the clinical syndrome, 32% of SHF-specific Dll4 heterozygotes demonstrate foreshortened and misaligned OFT, resulting in a double outlet right ventricle. Our murine model provides a molecular mechanism to explain the cardiac defects observed in AOS and establishes a novel clinical role for Dll4-mediated Notch signaling in SHF progenitor biology.


Subject(s)
Haploinsufficiency , Limb Deformities, Congenital , Animals , Disease Models, Animal , Ectodermal Dysplasia , Haploinsufficiency/genetics , Humans , Ligands , Limb Deformities, Congenital/genetics , Mice , Scalp Dermatoses/congenital
10.
BMC Anesthesiol ; 21(1): 117, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33858352

ABSTRACT

BACKGROUND: Adams-Oliver syndrome is characterized by the combination of congenital scalp defects and terminal transverse limb defects. In some instances, cardiovascular malformations and orofacial malformations have been observed. Little is written with regards to the anesthetic management and airway concerns of patients with Adams-Oliver syndrome. CASE PRESENTATION: A five-year-old female with Adams-Oliver syndrome presented for repeat lower extremity surgery. Airway exam was significant for dysmorphic features, such as hypertelorism, deviated jaw, and retrognathia. Video laryngoscope was utilized for intubation due to the patients retrognathic jaw, cranial deformities, and facial dysmorphism. A vein finder with ultrasound guidance was needed to place the peripheral intravenous line due to her history of difficult intravenous access. The patient was successfully intubated with slight cricoid pressure applied to direct the endotracheal tube smoothly. Surgery and recovery were both unremarkable. CONCLUSIONS: Due to varying presentations of Adams-Oliver syndrome, anesthetic and airway management considerations should be carefully assessed prior to surgery. Anesthesiologists must take into consideration possible orofacial abnormalities that may make intubation difficult. Amniotic band syndrome and other limb defects could potentially impact intravenous access as well.


Subject(s)
Ectodermal Dysplasia , Limb Deformities, Congenital , Scalp Dermatoses/congenital , Airway Management , Child, Preschool , Female , Humans
11.
Medicine (Baltimore) ; 100(9): e24633, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33655927

ABSTRACT

RATIONALE: Familial exudative vitreoretinopathy (FEVR) is an inherited disorder, which is mostly reported to be associated with the mutation of genes involved in the Wnt signaling pathway related to ß-catenin. To the best of our knowledge, the involvement of Adams-Oliver syndrome (AOS) genes in FEVR patients have not been reported before. PATIENT CONCERNS: Two patients with FEVR presented with microcephaly. One of them showed slight scarring of the scalp vertex which is a typical manifestation of AOS. The whole exon sequencing confirmed the diagnosis of AOS with 2 AOS-gene mutations at DOCK6 and ARHGAP31. Further clinical examination revealed that their parents with the same mutations showed FEVR-like vascular anomalies. DIAGNOSIS: Both patients were diagnosed with AOS through whole exon sequencing, and they presented with some FEVR-like retinopathy including retinal detachment. INTERVENTIONS: Both patients received vitrectomy for tractional retinal detachment with proliferative vitreoretinopathy. During the follow-up, 1 patient received additional laser photocoagulation for tractional retinal detachment. OUTCOMES: The 2 patients remained stable in the latest follow up after the treatment. LESSONS: Microcephaly could be associated with some form of retinopathy. We proposed that mutation of DOCK6 and ARHGAP31 genes could be the possible cause of FEVR associated with microcephaly. Our study suggested that these genes may be candidate genes of FEVR.


Subject(s)
Ectodermal Dysplasia/genetics , Familial Exudative Vitreoretinopathies/genetics , GTPase-Activating Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Limb Deformities, Congenital/genetics , Microcephaly/genetics , Phosphoproteins/genetics , Scalp Dermatoses/congenital , Eye Diseases, Hereditary/genetics , Female , Humans , Infant , Male , Mutation/genetics , Retinal Detachment/genetics , Scalp Dermatoses/genetics
13.
J AAPOS ; 24(3): 186-188, 2020 06.
Article in English | MEDLINE | ID: mdl-32522703

ABSTRACT

Adams-Oliver syndrome (AOS) is a congenital condition characterized by aplasia cutis congenita of the scalp and transverse limb defects. Other clinical features reported in association with AOS include cardiac malformations, cutis marmorata telangiectatica congenita, prenatal complications, and ophthalmic abnormalities. Reported ophthalmic manifestations range from Peters anomaly-like findings and cataract formation to incomplete or abnormal retinal vasculature, optic nerve hypoplasia, and rod dystrophy. We report the novel case of a 3-month-old boy with AOS type 2 who was found to have bilateral progressive macular ischemia.


Subject(s)
Ectodermal Dysplasia , Limb Deformities, Congenital , Macular Degeneration , Scalp Dermatoses/congenital , Female , Humans , Infant , Male , Pregnancy , Scalp
14.
Ophthalmic Genet ; 41(4): 377-380, 2020 08.
Article in English | MEDLINE | ID: mdl-32498638

ABSTRACT

BACKGROUND: Adams-Oliver syndrome (AOS) is a rare, inherited multi-systemic malformation syndrome characterized by a combination of aplasia cutis congenita and transverse terminal limb defects along with variable involvement of the central nervous system, eyes, and cardiovascular system. AOS can be inherited as both autosomal-dominant and recessive traits. Pathogenic variants in the DOCK6, ARHGAP31, EOGT, RBPJ, DLL4, and NOTCH1 genes have been associated with AOS. PURPOSE: To report a novel homozygous variant in the DOCK6 gene associated with Adams-Oliver syndrome type 2. MATERIALS AND METHODS: Case report. RESULTS: We report a case of a 4-month-old male who presented with microcephaly, global developmental delay, truncal hypotonia, and limb reduction defects. Ophthalmic examination revealed bilateral nystagmus and retinal detachment with mild cataractous changes in addition to retrolental plaque in the left eye. Next generation sequencing analysis identified a novel homozygous frameshift likely pathogenic variant (c.1269_1285dup (p.Arg429Glnfs*32)) in the DOCK6 gene. The constellation of the clinical findings and the genetic mutation were consistent with a diagnosis of AOS type 2. CONCLUSION: The discovery of this new likely pathogenic variant enriches the genotypic spectrum of DOCK6 gene and contributes to genetic diagnosis and counseling of families with AOS. Neurologic and ocular findings appear to be consistent with AOS type 2 for which multidisciplinary clinical evaluation is crucial.


Subject(s)
Ectodermal Dysplasia/pathology , Guanine Nucleotide Exchange Factors/genetics , Limb Deformities, Congenital/pathology , Mutation , Scalp Dermatoses/congenital , Ectodermal Dysplasia/genetics , Humans , Infant , Limb Deformities, Congenital/genetics , Male , Phenotype , Prognosis , Scalp Dermatoses/genetics , Scalp Dermatoses/pathology
15.
DNA Cell Biol ; 39(5): 783-789, 2020 May.
Article in English | MEDLINE | ID: mdl-32129674

ABSTRACT

Adams-Oliver syndrome (AOS) is a rare hereditary disorder characterized by aplasia cutis congenita (ACC) and terminal transverse limb defects. The etiology of AOS has remained largely unknown, although mutations in the notch receptor 1 (NOTCH1) gene are most common genetic alteration associated with this disease. In this study, we aimed to identify the case of a 6-year-old boy, who presented with large ACC of the scalp and aortic valve stenosis, suggesting the possibility of AOS. Whole-exome sequencing identified a novel, de novo, in-frame deletion in the NOTCH1 gene (NOTCH1 c.1292_1294del, p.Asn431del) in the patient. The p.Asn431del variant was evaluated by several in silico analyses, which predicted that the mutant was likely to be pathogenic. In addition, molecular modeling with the PyMOL Molecular Graphics System suggested that the NOTCH1-N431del destabilizes calcium ion chelation, leading to decreased receptor-ligand binding efficiency. Quantitative reverse transcription PCR showed further significant downregulation of the Notch target genes, hes-related family bHLH transcription factor with YRPW motif 1 (HEY1) and hes family bHLH transcription factor 1 (HES1), suggesting that this mutation causes disease through dysregulation of the Notch signaling pathway. Our study provides evidence that the NOTCH1-N431del mutation is responsible for this case of AOS. To our knowledge, this is the first report of a patient with AOS caused by NOTCH1 mutation in Asia, and this information will be useful for providing the family with genetic counseling that can help to guide their future plans.


Subject(s)
Ectodermal Dysplasia/genetics , Frameshift Mutation , Limb Deformities, Congenital/genetics , Receptor, Notch1/genetics , Scalp Dermatoses/congenital , Amino Acid Sequence , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Child , China , Humans , Male , Models, Molecular , Protein Conformation , Receptor, Notch1/chemistry , Repressor Proteins/genetics , Scalp Dermatoses/genetics , Transcription Factor HES-1/genetics
16.
Am J Med Genet A ; 182(1): 29-37, 2020 01.
Article in English | MEDLINE | ID: mdl-31654484

ABSTRACT

RATIONALE: Adams-Oliver syndrome (AOS) is a genetic disorder characterized by the association of aplasia cutis congenita (ACC), terminal transverse limb defect (TTLD), congenital cardiac malformation (CCM), and minor features, such as cutaneous, neurological, and hepatic abnormalities (HAs). The aim of the study is to emphasize phenotype-genotype correlations in AOS. METHODS: We studied 29 AOS patients. We recorded retrospectively detailed phenotype data, including clinical examination, biological analyses, and imaging. The molecular analysis was performed through whole exome sequencing (WES). RESULTS: Twenty-nine patients (100%) presented with ACC, the principal inclusion criteria in the study. Seventeen of twenty-one (81%) had cutis marmorata telangiectasia congenita, 16/26 (62%) had TTLD, 14/23 (61%) had CCM, 7/20 (35%) had HAs, and 9/27 (33%) had neurological findings. WES was performed in 25 patients. Fourteen of twenty-five (56%) had alterations in the genes already described in AOS. CCM and HAs are particularly associated with the NOTCH1 genotype. TTLD is present in patients with DOCK6 and EOGT alterations. Neurological findings of variable degree were associated sometimes with DOCK6 and NOTCH1 rarely with EOGT. CONCLUSION: AOS is characterized by a clinical and molecular variability. It appears that degrees of genotype-phenotype correlations exist for patients with identified pathogenic mutations, underlining the need to undertake a systematic but adjusted multidisciplinary assessment.


Subject(s)
Ectodermal Dysplasia/genetics , Genetic Predisposition to Disease , Guanine Nucleotide Exchange Factors/genetics , Limb Deformities, Congenital/genetics , Receptor, Notch1/genetics , Scalp Dermatoses/congenital , Amniotic Band Syndrome/genetics , Amniotic Band Syndrome/pathology , Ectodermal Dysplasia/etiology , Ectodermal Dysplasia/pathology , Female , Genetic Association Studies , Genotype , Humans , Limb Deformities, Congenital/etiology , Limb Deformities, Congenital/pathology , Liver Diseases/genetics , Liver Diseases/pathology , Male , Mutation/genetics , Pedigree , Phenotype , Scalp Dermatoses/etiology , Scalp Dermatoses/genetics , Scalp Dermatoses/pathology , Upper Extremity Deformities, Congenital/genetics , Upper Extremity Deformities, Congenital/pathology , Exome Sequencing
18.
Am J Med Genet A ; 179(11): 2246-2251, 2019 11.
Article in English | MEDLINE | ID: mdl-31368252

ABSTRACT

Adams-Oliver syndrome (AOS) is a rare congenital disease characterized by aplasia cutis congenita (ACC) and terminal transverse limb defects (TTLD). It shows significant genetic heterogeneity and can be transmitted by autosomal dominant or recessive inheritance. Recessive inheritance is associated with mutations of DOCK6 or EOGT; however, only few cases have been published so far. We present two families with EOGT-associated AOS. Due to pseudodominance in one family, the recognition of the recessive inheritance pattern was difficult. We identified two novel AOS-causing mutations (c.404G>A/p.Cys135Tyr and c.311+1G>T). The phenotype in the presented families was dominated by large ACC, whereas TTLD were mostly subtle or even absent and no major malformations occured. Our observations along with the previously published cases indicate that the two types of recessive AOS (EOGT- vs. DOCK6-associated) differ significanty regarding the frequency of neurologic or ocular deficits.


Subject(s)
Ectodermal Dysplasia/diagnosis , Ectodermal Dysplasia/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Limb Deformities, Congenital/diagnosis , Limb Deformities, Congenital/genetics , Mutation , N-Acetylglucosaminyltransferases/genetics , Scalp Dermatoses/congenital , Child , Consanguinity , Exons , Genetic Association Studies/methods , Humans , Infant , Magnetic Resonance Imaging , Male , Pedigree , Phenotype , Scalp Dermatoses/diagnosis , Scalp Dermatoses/genetics
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(4): 348-351, 2019 Apr 10.
Article in Chinese | MEDLINE | ID: mdl-30950023

ABSTRACT

OBJECTIVE: To detect pathogenic mutation of DOCK6 gene in a patient with convulsive seizure and refractory epilepsy. METHODS: CytoScan HD-Array and next generation sequencing were used to detect the potential mutation in the patient. RESULTS: The proband has carried compound heterozygous mutations of c.188C>T (p.Arg63Gln) and c.5374C>T (p.Glu1792Lys) of the DOCK6 gene, which were respectively inherited from his mother and father. Neither mutation was reported previously. Bioinformatic analysis indicated that the two amino acids are highly conserved. Based on the ACMG guidelines, the c.188C>T mutation was predicted to be likely pathogenic, while the c.5374C>T mutation was of uncertain significance. CONCLUSION: The compound heterozygous mutations of c.188C>T (p.Arg63Gln) and c.5374C>T (p.Glu1792Lys) of the DOCK6 gene probably underlie the disease in this patient.


Subject(s)
Ectodermal Dysplasia/genetics , Guanine Nucleotide Exchange Factors/genetics , Limb Deformities, Congenital/genetics , Scalp Dermatoses/congenital , Child , Humans , Mutation , Pedigree , Scalp Dermatoses/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...