Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Am J Ophthalmol Case Rep ; 36: 102091, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39027323

ABSTRACT

Purpose: To describe unique ocular features in a child with Joubert syndrome type 6. Observations: A 4-year-old male patient presented with right microphthalmia and non-dilating pupil and left primary position nystagmus. Brain MRI revealed a "molar tooth sign" of the midbrain and a "batwing sign" of the fourth ventricle along with large retroorbital cysts bilaterally. The diagnosis of autosomal recessive Joubert syndrome type 6 due to homozygous pathogenic variant c.725A > G p. (Asn242Ser) in TMEM67 gene was confirmed by whole exome sequencing. Left eye had nystagmus and the left optic nerve and retina showed epipapillary and subretinal fibrosis, respectively. Scleral buckle was performed for left non-rhegmatogenous retinal detachment which then improved and has been stable. Conclusions and Importance: We present a rare case of JS with some unique ophthalmic features which expand clinical knowledge on this complex systemic and ocular entity.

2.
BMC Med Genomics ; 17(1): 156, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844949

ABSTRACT

BACKGROUND: Recurrent pregnancy loss (RPL) is a common pregnancy complication that brings great pain to pregnant women and their families. Genetic factors are an important cause reason of RPL. However, clinical research on monogenic diseases with recurrent miscarriage is insufficient. CASE PRESENTATION: Here we reported a Chinese family with RPL and genetic analysis of the abortion and parents. A paternally inherited heterozygous missense variant c.1415T > G (p.V472G) and a maternally inherited heterozygous nonsense variant c.2314del (p.M772*) in TMEM67 gene were identified by trio-exome sequencing. c.2314del (p.M772*) generated a premature stop codon and truncated protein, was classified as "pathogenic". c.1415T > G (p.V472G) located in extra-cellular region, was classified as "likely pathogenic". Biallelic variants in TMEM67 gene cause lethal Meckel syndrome 3, consistent with the proband's prenatal phenotype. CONCLUSION: The current study of the Chinese family expands the pathogenic variant spectrum of TMEM67 and emphasizes the necessity of exome sequencing in RPL condition.


Subject(s)
Abortion, Habitual , Membrane Proteins , Pedigree , Humans , Female , Membrane Proteins/genetics , Abortion, Habitual/genetics , Pregnancy , Adult , Asian People/genetics , Male , Exome Sequencing , China , East Asian People
3.
Nephron ; 148(4): 264-272, 2024.
Article in English | MEDLINE | ID: mdl-36617405

ABSTRACT

The clinical features of cerebellar vermis hypoplasia, oligophrenia, ataxia, coloboma, and hepatic fibrosis (COACH) characterize the rare autosomal recessive multisystem disorder called COACH syndrome. COACH syndrome belongs to the spectrum of Joubert syndrome and related disorders (JSRDs) and liver involvement distinguishes COACH syndrome from the rest of the JSRD spectrum. Developmental delay and oculomotor apraxia occur early but with time, these can improve and may not be readily apparent or no longer need active medical management. Congenital hepatic fibrosis and renal disease, on the other hand, may develop late, and the temporal incongruity in organ system involvement may delay the recognition of COACH syndrome. We present a case of a young adult presenting late to a Renal Genetics Clinic for evaluation of renal cystic disease with congenital hepatic fibrosis, clinically suspected to have autosomal recessive polycystic kidney disease. Following genetic testing, a reevaluation of his medical records from infancy, together with reverse phenotyping and genetic phasing, led to a diagnosis of COACH syndrome.


Subject(s)
Abnormalities, Multiple , Brain/abnormalities , Cerebellar Vermis , Cerebellum/abnormalities , Cholestasis , Coloboma , Genetic Diseases, Inborn , Intellectual Disability , Liver Diseases , Nervous System Malformations , Polycystic Kidney, Autosomal Recessive , Young Adult , Humans , Coloboma/diagnosis , Coloboma/genetics , Polycystic Kidney, Autosomal Recessive/diagnosis , Polycystic Kidney, Autosomal Recessive/genetics , Delayed Diagnosis , Genotype , Liver Cirrhosis/genetics , Ataxia/diagnosis , Ataxia/genetics , Intellectual Disability/genetics , Developmental Disabilities
5.
Biochem Biophys Res Commun ; 636(Pt 1): 162-169, 2022 12 25.
Article in English | MEDLINE | ID: mdl-36334440

ABSTRACT

Primary cilia transduce signals via transmembrane and membrane-associated proteins localized to the ciliary membrane in vertebrate cells. In humans, transmembrane protein 67 (TMEM67), a component of the multiprotein complex functioning as a gatekeeper at the transition zone (TZ) of primary cilia, is mutated in patients suffering from cilia-related pleiotropic diseases, collectively referred to as ciliopathies. The requirement of TMEM67 for the gating function of the TZ that delivers membrane proteins into the ciliary compartment has not been determined. In this study, we established hTERT-RPE1 cells with knockout (KO) of TMEM67 and examined whether cilium formation and TZ gating are affected by its ablation. TMEM67-KO cells displayed impaired ciliogenesis, elongated cilia, perturbed ciliary localization of membrane-associated proteins ARL13B and INPP5E but normal recruitment of TZ proteins CEP290, RPGRIP1L and NPHP5. The exogenous expression of ciliopathy-associated TMEM67 mutants restored ciliary localization of ARL13B and INPP5E but failed to attenuate aberrant cilium elongation in TMEM67-KO cells. Furthermore, we found that TMEM67 localization is not confined to the TZ but extends into the cilium. Our findings indicate that TMEM67 is required not only for ciliogenesis and cilium length regulation but also for the gating function of the TZ independently of RPGRIP1L/CEP290/NPHP5 recruitment to this region. They further suggest that aberrant cilium elongation underlies the pathogenesis of TMEM67-linked ciliopathies.


Subject(s)
Cilia , Ciliopathies , Humans , Cilia/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Antigens, Neoplasm/metabolism , Cytoskeletal Proteins/metabolism , Cell Cycle Proteins/metabolism , ADP-Ribosylation Factors/metabolism
6.
J Cell Physiol ; 237(6): 2713-2723, 2022 06.
Article in English | MEDLINE | ID: mdl-35621037

ABSTRACT

TMEM67 (mecklin or MKS3) locates in the transition zone of cilia. Dysfunction of TMEM67 disrupts cilia-related signaling and leads to developmental defects of multiple organs in humans. Typical autosomal recessive TMEM67 defects cause partial overlapping phenotypes, including abnormalities in the brain, eyes, liver, kidneys, bones, and so forth. However, emerging reports of isolated nephronophthisis suggest the possibility of a broader phenotype spectrum. In this study, we analyzed the genetic data of cholestasis patients with no obvious extrahepatic involvement but with an unexplained high level of gamma-glutamyl transpeptidase (GGT). We identified five Han Chinese patients from three unrelated families with biallelic nonnull low-frequency TMEM67 variants. All variants were predicted pathogenic in silico, of which p. Arg820Ile and p. Leu144del were previously unreported. In vitro studies revealed that the protein levels of the TMEM67 variants were significantly decreased; however, their interaction with MKS1 remained unaffected. All the patients, aged 7-39 years old, had silently progressive cholestasis with elevated GGT but had normal bilirubin levels. Histological studies of liver biopsy of patients 1, 3, and 5 showed the presence of congenital hepatic fibrosis. We conclude that variants in TMEM67 are associated with a mild phenotype of unexplained, persistent, anicteric, and high GGT cholestasis without typical symptoms of TMEM67 defects; this possibility should be considered by physicians in gastroenterology and hepatology.


Subject(s)
Cholestasis , gamma-Glutamyltransferase , Cholestasis/genetics , Genetic Diseases, Inborn , Humans , Liver Cirrhosis/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phenotype , gamma-Glutamyltransferase/genetics
7.
Genes (Basel) ; 12(7)2021 07 16.
Article in English | MEDLINE | ID: mdl-34356094

ABSTRACT

Renal cystic diseases are characterized by genetic and phenotypic heterogeneity. Congenital renal cysts can be classified as developmental disorders and are commonly diagnosed prenatally using ultrasonography and magnetic resonance imaging. Progress in molecular diagnostics and availability of exome sequencing procedures allows diagnosis of single-gene disorders in the prenatal period. Two patients with a prenatal diagnosis of polycystic kidney disease are presented in this article. TMEM67 mutations were identified in both fetuses using a whole-exome sequencing (WES) study. In one of them, the phenotypic syndrome diagnosed prenatally was different from that diagnosed in the postnatal period.


Subject(s)
Abnormalities, Multiple/diagnosis , Cerebellum/abnormalities , Ciliary Motility Disorders/diagnosis , Encephalocele/diagnosis , Eye Abnormalities/diagnosis , Kidney Diseases, Cystic/diagnosis , Membrane Proteins/genetics , Polycystic Kidney Diseases/diagnosis , Retina/abnormalities , Retinitis Pigmentosa/diagnosis , Abnormalities, Multiple/genetics , Ciliary Motility Disorders/genetics , Diagnosis, Differential , Encephalocele/genetics , Eye Abnormalities/genetics , Female , Humans , Kidney Diseases, Cystic/genetics , Membrane Proteins/metabolism , Mutation , Polycystic Kidney Diseases/genetics , Pregnancy , Prenatal Diagnosis/methods , Retinitis Pigmentosa/genetics , Exome Sequencing/methods
8.
J Cell Mol Med ; 2021 May 25.
Article in English | MEDLINE | ID: mdl-34032358

ABSTRACT

Polycystic kidney disease (PKD) is known to occur in three main forms, namely autosomal dominant PKD (ADPKD), autosomal recessive PKD (ARPKD) and syndromic PKD (SPKD), based on the clinical manifestations and genetic causes, which are diagnosable from the embryo stage to the later stages of life. Selection of the genetic test for the individuals with diagnostic imaging reports of cystic kidneys without a family history of the disease continues to be a challenge in clinical practice. With the objective of maintaining a limit on the time and medical cost of the procedure, a practical strategy for genotyping and targeted validation to resolve cystogene variations was developed in our clinical laboratory, which combined the techniques of whole-exome sequencing (WES), Long-range PCR (LR-PCR), Sanger sequencing and multiplex ligation-dependent probe amplification (MLPA) to work in a stepwise approach. In this context, twenty-six families with renal polycystic disorders were enrolled in the present study. Thirty-two variants involving four ciliary genes (PKD1, PKHD1, TMEM67 and TMEM107) were identified and verified in 23 families (88.5%, 23/26), which expanded the variant spectrum by 16 novel variants. Pathogenic variations in five foetuses of six families diagnosed with PKD were identified using prenatal ultrasound imaging. Constitutional biallelic and digenic variations constituted the pathogenic patterns in these foetuses. The preliminary clinical data highlighted that the WES + LR PCR-based workflow followed in the present study is efficient in detecting divergent variations in PKD. The biallelic and digenic mutations were revealed as the main pathogenic patterns in the foetuses with PKD.

9.
BMC Med Genet ; 21(1): 18, 2020 01 30.
Article in English | MEDLINE | ID: mdl-32000717

ABSTRACT

BACKGROUND: Joubert syndrome is a genetically heterogeneous autosomal recessive ciliopathy characterized by the combination of hypoplasia/aplasia of the cerebellar vermis, thickened and elongated superior cerebellar peduncles and a deep interpeduncular fossa, known as "molar tooth sign" associated with hypotonia, respiratory control disturbances and abnormal eye movements. To date, pathogenic variants in over 35 genes are known to cause autosomal recessive Joubert Syndrome, while one gene is associated with X-linked recessive inheritance. CASE PRESENTATION: We describe here a non-consanguineous Vietnamese family with Joubert syndrome, a fetus and 10-year-old developmentally delayed boy. Ultrasonography showed ventriculomegaly at 26 + 6 weeks of gestation in the fetus. The 10-year-old-boy was diagnosed with cerebral palsy of unknown origin. Clinical physical examination at the age of 10, he showed clinical features of Joubert syndrome including typical facial dysmorphism, ataxia, severe psychomotor delay, oculomotor apraxia and molar tooth sign on brain MRI. Whole exome sequencing analysis identified a novel compound heterozygous c.725A > G p.Asn242Ser and c.313-3 T > G p.Lys105Valfs*16 TMEM67 variant in the proband and the affected fetus. These two variants were inherited from each parent and confirmed by Sanger sequencing. The variant c.725A > G p.Asn242Ser was previously documented in patients with JS, the novel splice-site c.313-3 T > G p.Lys105Valfs*16 TMEM67 variant produced an aberrant transcript with the loss of four nucleotides of exon 03. CONCLUSION: This study confirms the diagnosis of Joubert syndrome in a Vietnamese family and expands the mutational spectrum of TMEM67 sequence variations. We also highlight the importance of molecular approaches to unravel underlying mechanisms of human genetic disorders. Early precise diagnosis could help provide further accurate genetic counseling for recurrence-risk assessment, future diagnostic option, management as well as treatment guidance for rare disorders.


Subject(s)
Abnormalities, Multiple/genetics , Cerebellum/abnormalities , Eye Abnormalities/genetics , Genetic Predisposition to Disease , Kidney Diseases, Cystic/genetics , Membrane Proteins/genetics , Retina/abnormalities , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/physiopathology , Adult , Cerebellum/physiopathology , Child , Exons/genetics , Eye Abnormalities/diagnosis , Eye Abnormalities/physiopathology , Female , Humans , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/physiopathology , Magnetic Resonance Imaging , Male , Mutation/genetics , Pedigree , Protein Isoforms/genetics , Retina/metabolism , Retina/pathology , Retina/physiopathology , Vietnam , Exome Sequencing
10.
Clin Genet ; 96(6): 560-565, 2019 12.
Article in English | MEDLINE | ID: mdl-31411728

ABSTRACT

Meckel syndrome (MKS) is a perinatally lethal, genetically heterogeneous, autosomal recessive condition caused by defective primary cilium formation leading to polydactyly, multiple cysts in kidneys and malformations of nervous system. We performed exome sequencing in six fetuses from six unrelated families with MKS. We identified seven novel variants in B9D2, TNXDC15, CC2D2A, CEP290 and TMEM67. We describe the second family with MKS due to a homozygous variant in B9D2 and fifth family with bi-allelic variant in TXNDC15. Our data validates the causation of MKS by pathogenic variation in B9D2 and TXNDC15 and also adds novel variants in CC2D2A, CEP290 and TMEM67 to the literature.


Subject(s)
Ciliary Motility Disorders/genetics , Ciliary Motility Disorders/pathology , Encephalocele/genetics , Encephalocele/pathology , Fetus/abnormalities , Mutation/genetics , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Amino Acid Sequence , Base Sequence , Chromosome Segregation/genetics , Ciliary Motility Disorders/diagnostic imaging , Cohort Studies , Encephalocele/diagnostic imaging , Female , Humans , Male , Pedigree , Phenotype , Polycystic Kidney Diseases/diagnostic imaging , Retinitis Pigmentosa/diagnostic imaging
11.
Clin Case Rep ; 6(11): 2189-2192, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30455918

ABSTRACT

We present a patient with a clinical diagnosis of Joubert syndrome with COACH phenotype who carries two TMEM67 variants of uncertain significance (VUS). One VUS can be reclassified as "likely pathogenic" by adding clinical data. As genetic testing becomes more accessible, more VUS will require clinical correlation for accurate classification.

12.
Public Health Genomics ; 20(3): 188-193, 2017.
Article in English | MEDLINE | ID: mdl-28719906

ABSTRACT

BACKGROUND: Joubert syndrome (JS) is a clinically and genetically heterogeneous group of rare neurodevelopmental disorder characterised by peculiar midbrain-hindbrain malformation, known as the "molar tooth" sign. JS can manifest a broad range of signs and symptoms. The most common features of JS are hypotonia, ataxia, developmental delay/intellectual disability, abnormal eye movements, and neonatal breathing abnormalities. To date, 29 genes have been shown to cause JS. METHODS: We employed whole-genome single nucleotide polymorphism genotyping in a group of Iranian families with JS and Sanger sequencing of a known mutation associated with JS located in a single homozygous regions shared by affected members of the families. RESULTS: Homozygosity mapping uncovered a shared ∼2.2-Mb run of homozygosity on chromosome 8q21.3-q22.1 encompassing the known JS-causing TMEM67 gene. Sanger sequencing of a known mutation (NM_153704.5: c.725A>G; p.Asn242Ser) in TMEM67 identified from studying another Iranian family using whole-exome sequencing confirmed the presence of the homozygous mutation in 22 affected members of 12 nuclear families. "Molar tooth" sign of brain magnetic resonance imaging, moderate-to-severe neurodevelopmental delay, and abnormal eye movements were the most common features of affected individuals. In addition, liver disease, seizure, behavioural abnormalities, failure to thrive, and kidney disease were observed variably in some of the patients. CONCLUSION: We propose that Asn242Ser is a founder mutation in the Iranian population, which might explain a significant proportion of JS cases from eastern Iran. Therefore, screening for this variant should be considered for genetic testing in Iranian patients with JS. In addition, this finding is important for developing population-specific genetic testing in Iran.


Subject(s)
Abnormalities, Multiple/genetics , Cerebellum/abnormalities , Eye Abnormalities/genetics , Founder Effect , Kidney Diseases, Cystic/genetics , Membrane Proteins/genetics , Mutation/genetics , Retina/abnormalities , Failure to Thrive/genetics , Female , Genetic Testing , Genome-Wide Association Study , Homozygote , Humans , Iran , Liver Diseases/genetics , Magnetic Resonance Imaging , Male , Pedigree , Polymorphism, Single Nucleotide/genetics , Young Adult
13.
Clin Case Rep ; 5(7): 1098-1102, 2017 07.
Article in English | MEDLINE | ID: mdl-28680603

ABSTRACT

We report an otherwise healthy 32-year-old man with portal hypertension, variceal bleeding, and congenital hepatic fibrosis with ductal plate malformation. Genetic screening identified two TMEM67 mutations. Biallelic TMEM67 mutations are known to cause Joubert/Meckel syndrome or nephronopthisis with hepatic fibrosis, but have never been found in isolated hepatic fibrosis.

14.
Pediatr Nephrol ; 32(10): 1989-1992, 2017 10.
Article in English | MEDLINE | ID: mdl-28620746

ABSTRACT

BACKGROUND: Meckel-Gruber syndrome (MKS, OMIM #607361) is a rare pre- or perinatal lethal autosomal recessive ciliopathy caused by mutations in at least 12 known genes. It has a clinical and genetic overlap with other viable ciliopathies, especially Joubert syndrome and Joubert syndrome-related disorders. MKS is characterized by multicystic kidney dysplasia, central nervous system malformations (usually occipital encephalocele), ductal plate malformation of the liver, and postaxial polydactyly. CASE DIAGNOSIS: We identified a homozygous mutation in TMEM67 (MKS3) in a fetus affected by MKS; however, only the mother was a carrier of the respective mutation. Genotyping with polymorphic microsatellite markers and single nucleotide polymorphism (SNP) array revealed a maternal uniparental disomy (UPD) of the entire chromosome 8 (upd(8)mat), harboring TMEM67. CONCLUSIONS: This is the first reported case of UPD as a cause of MKS. The possible underlying mechanisms for uniparental disomy (UPD) are reviewed. Even if rare, awareness of UPD and comprehensive work-up in the case of unexpected homozygosity for a recessive mutation is essential for accurate genetic counseling and assessment of the risk of recurrence.


Subject(s)
Chromosomes, Human, Pair 8/genetics , Ciliary Motility Disorders/genetics , Encephalocele/genetics , Fetal Diseases/genetics , Membrane Proteins/genetics , Polycystic Kidney Diseases/genetics , Retinitis Pigmentosa/genetics , Uniparental Disomy , Abortion, Induced , Adult , Ciliary Motility Disorders/diagnosis , DNA Mutational Analysis/methods , Encephalocele/diagnosis , Female , Fetal Diseases/diagnosis , Genetic Testing/methods , Homozygote , Humans , Karyotyping/methods , Male , Mutation , Polycystic Kidney Diseases/diagnosis , Pregnancy , Prenatal Diagnosis , Retinitis Pigmentosa/diagnosis , Ultrasonography, Prenatal
15.
Int J Clin Exp Pathol ; 8(5): 5379-86, 2015.
Article in English | MEDLINE | ID: mdl-26191240

ABSTRACT

Meckel-Gruber syndrome (MKS) is a lethal autosomal recessive condition characterized by renal cysts and variably associated features, including developmental anomalies of the central nervous system (typically encephalocele), hepatic ductal dysplasia and cysts, and polydactyly. Genetic heterogeneity has been demonstrated at eleven loci, MKS1-11. Here, we present the clinical and molecular characteristics of a Chinese MKS3 family with occipital encephalocele and kidney enlargement. DNA sequencing of affected fetuses revealed a homozygous c.1645C>T substitution in exon 16 of TMEM67, leading to a p.R549C substitution in meckelin. The R549 residue is highly conserved across human, rat, mouse, zebrafish, chicken, wolf and platypus genomes. Hha I restriction analysis demonstrated that the c.1645C>T mutation was absent in 200 unrelated control chromosomes of Chinese background, supporting the hypothesis that it represents causative mutation, not rare polymorphism. Our data provide additional molecular and clinical information for establishing a better genotype-phenotype understanding of MKS.


Subject(s)
Ciliary Motility Disorders/genetics , Encephalocele/genetics , Membrane Proteins/genetics , Mutation, Missense , Polycystic Kidney Diseases/genetics , Abnormalities, Multiple/genetics , China , DNA Mutational Analysis , Family , Humans
16.
Dis Model Mech ; 8(6): 527-41, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26035863

ABSTRACT

Ciliopathies are a group of developmental disorders that manifest with multi-organ anomalies. Mutations in TMEM67 (MKS3) cause a range of human ciliopathies, including Meckel-Gruber and Joubert syndromes. In this study we describe multi-organ developmental abnormalities in the Tmem67(tm1Dgen/H1) knockout mouse that closely resemble those seen in Wnt5a and Ror2 knockout mice. These include pulmonary hypoplasia, ventricular septal defects, shortening of the body longitudinal axis, limb abnormalities, and cochlear hair cell stereociliary bundle orientation and basal body/kinocilium positioning defects. The basal body/kinocilium complex was often uncoupled from the hair bundle, suggesting aberrant basal body migration, although planar cell polarity and apical planar asymmetry in the organ of Corti were normal. TMEM67 (meckelin) is essential for phosphorylation of the non-canonical Wnt receptor ROR2 (receptor-tyrosine-kinase-like orphan receptor 2) upon stimulation with Wnt5a-conditioned medium. ROR2 also colocalises and interacts with TMEM67 at the ciliary transition zone. Additionally, the extracellular N-terminal domain of TMEM67 preferentially binds to Wnt5a in an in vitro binding assay. Cultured lungs of Tmem67 mutant mice failed to respond to stimulation of epithelial branching morphogenesis by Wnt5a. Wnt5a also inhibited both the Shh and canonical Wnt/ß-catenin signalling pathways in wild-type embryonic lung. Pulmonary hypoplasia phenotypes, including loss of correct epithelial branching morphogenesis and cell polarity, were rescued by stimulating the non-canonical Wnt pathway downstream of the Wnt5a-TMEM67-ROR2 axis by activating RhoA. We propose that TMEM67 is a receptor that has a main role in non-canonical Wnt signalling, mediated by Wnt5a and ROR2, and normally represses Shh signalling. Downstream therapeutic targeting of the Wnt5a-TMEM67-ROR2 axis might, therefore, reduce or prevent pulmonary hypoplasia in ciliopathies and other congenital conditions.


Subject(s)
Body Patterning , Ciliary Motility Disorders/metabolism , Encephalocele/metabolism , Epithelium/embryology , Membrane Proteins/metabolism , Morphogenesis , Polycystic Kidney Diseases/metabolism , Wnt Signaling Pathway , Animals , Animals, Newborn , Cell Differentiation , Cell Polarity , Cilia/metabolism , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Epithelium/metabolism , HEK293 Cells , Humans , Lung/embryology , Lung/metabolism , Membrane Proteins/deficiency , Mice , Mutation/genetics , Organ of Corti/abnormalities , Organ of Corti/embryology , Organ of Corti/pathology , Phenotype , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Retinitis Pigmentosa , Stereocilia/metabolism , Wnt Proteins/metabolism , Wnt-5a Protein , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL