Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Front Endocrinol (Lausanne) ; 14: 1231828, 2023.
Article in English | MEDLINE | ID: mdl-37964950

ABSTRACT

Noonan, Costello and Cardio-facio-cutaneous syndromes belong to a group of disorders named RASopathies due to their common pathogenetic origin that lies on the Ras/MAPK signaling pathway. Genetics has eased, at least in part, the distinction of these entities as they are presented with overlapping clinical features which, sometimes, become more pronounced with age. Distinctive face, cardiac and skeletal defects are among the primary abnormalities seen in these patients. Skeletal dysmorphisms range from mild to severe and may include anterior chest wall anomalies, scoliosis, kyphosis, short stature, hand anomalies, muscle weakness, osteopenia or/and osteoporosis. Patients usually have increased serum concentrations of bone resorption markers, while markers of bone formation are within normal range. The causative molecular defects encompass the members of the Ras/MAPK/ERK pathway and the adjacent cascades, important for the maintenance of normal bone homeostasis. It has been suggested that modulation of the expression of specific molecules involved in the processes of bone remodeling may affect the osteogenic fate decision, potentially, bringing out new pharmaceutical targets. Currently, the laboratory imprint of bone metabolism on the clinical picture of the affected individuals is not clear, maybe due to the rarity of these syndromes, the small number of the recruited patients and the methods used for the description of their clinical and biochemical profiles.


Subject(s)
Ectodermal Dysplasia , Heart Defects, Congenital , Humans , ras Proteins/metabolism , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Failure to Thrive
2.
Hum Mol Genet ; 31(15): 2535-2547, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35220430

ABSTRACT

Epidermal development and maintenance are finely regulated events requiring a strict balance between proliferation and differentiation. Alterations in these processes give rise to human disorders such as cancer or syndromes with skin and annexes defects, known as ectodermal dysplasias (EDs). Here, we studied the functional effects of two novel receptor-interacting protein kinase 4 (RIPK4) missense mutations identified in siblings with an autosomal recessive ED with cutaneous syndactyly, palmoplantar hyperkeratosis and orofacial synechiae. Clinical overlap with distinct EDs caused by mutations in transcription factors (i.e. p63 and interferon regulatory factor 6, IRF6) or nectin adhesion molecules was noticed. Impaired activity of the RIPK4 kinase resulted both in altered epithelial differentiation and defective cell adhesion. We showed that mutant RIPK4 resulted in loss of PVRL4/nectin-4 expression in patient epidermis and primary keratinocytes, and demonstrated that PVRL4 is transcriptionally regulated by IRF6, a RIPK4 phosphorylation target. In addition, defective RIPK4 altered desmosome morphology through modulation of plakophilin-1 and desmoplakin. In conclusion, this work implicates RIPK4 kinase function in the p63-IRF6 regulatory loop that controls the proliferation/differentiation switch and cell adhesion, with implications in ectodermal development and cancer.


Subject(s)
Ectodermal Dysplasia , Interferon Regulatory Factors , Cell Adhesion/genetics , Cell Adhesion Molecules/metabolism , Ectodermal Dysplasia/metabolism , Homeostasis , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Keratinocytes/metabolism , Nectins , Protein Serine-Threonine Kinases
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34548398

ABSTRACT

Skeletal ciliopathies (e.g., Jeune syndrome, short rib polydactyly syndrome, and Sensenbrenner syndrome) are frequently associated with nephronophthisis-like cystic kidney disease and other organ manifestations. Despite recent progress in genetic mapping of causative loci, a common molecular mechanism of cartilage defects and cystic kidneys has remained elusive. Targeting two ciliary chondrodysplasia loci (ift80 and ift172) by CRISPR/Cas9 mutagenesis, we established models for skeletal ciliopathies in Xenopus tropicalis Froglets exhibited severe limb deformities, polydactyly, and cystic kidneys, closely matching the phenotype of affected patients. A data mining-based in silico screen found ttc30a to be related to known skeletal ciliopathy genes. CRISPR/Cas9 targeting replicated limb malformations and renal cysts identical to the models of established disease genes. Loss of Ttc30a impaired embryonic renal excretion and ciliogenesis because of altered posttranslational tubulin acetylation, glycylation, and defective axoneme compartmentalization. Ttc30a/b transcripts are enriched in chondrocytes and osteocytes of single-cell RNA-sequenced embryonic mouse limbs. We identify TTC30A/B as an essential node in the network of ciliary chondrodysplasia and nephronophthisis-like disease proteins and suggest that tubulin modifications and cilia segmentation contribute to skeletal and renal ciliopathy manifestations of ciliopathies in a cell type-specific manner. These findings have implications for potential therapeutic strategies.


Subject(s)
Bone and Bones/abnormalities , Ciliopathies/pathology , Craniosynostoses/pathology , Cytoskeletal Proteins/metabolism , Ectodermal Dysplasia/pathology , Embryo, Nonmammalian/pathology , Musculoskeletal Abnormalities/pathology , Polycystic Kidney Diseases/pathology , Tubulin/chemistry , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Ciliopathies/genetics , Ciliopathies/metabolism , Craniosynostoses/genetics , Craniosynostoses/metabolism , Cytoskeletal Proteins/genetics , Disease Models, Animal , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Embryo, Nonmammalian/metabolism , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/metabolism , Phenotype , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Tubulin/metabolism , Xenopus laevis
4.
Dev Dyn ; 250(8): 1074-1095, 2021 08.
Article in English | MEDLINE | ID: mdl-33522658

ABSTRACT

BACKGROUND: Cardio-facio-cutaneous (CFC) syndrome is a human multiple congenital anomaly syndrome that is caused by activating heterozygous mutations in either BRAF, MEK1, or MEK2, three protein kinases of the Ras/mitogen-activated protein kinase (MAPK) pathway. CFC belongs to a group of syndromes known as RASopathies. Skeletal muscle hypotonia is a ubiquitous phenotype of RASopathies, especially in CFC syndrome. To better understand the underlying mechanisms for the skeletal myopathy in CFC, a mouse model with an activating BrafL597V allele was utilized. RESULTS: The activating BrafL597V allele resulted in phenotypic alterations in skeletal muscle characterized by a reduction in fiber size which leads to a reduction in muscle size which are functionally weaker. MAPK pathway activation caused inhibition of myofiber differentiation during embryonic myogenesis and global transcriptional dysregulation of developmental pathways. Inhibition in differentiation can be rescued by MEK inhibition. CONCLUSIONS: A skeletal myopathy was identified in the CFC BrafL597V mouse validating the use of models to study the effect of Ras/MAPK dysregulation on skeletal myogenesis. RASopathies present a novel opportunity to identify new paradigms of myogenesis and further our understanding of Ras in development. Rescue of the phenotype by inhibitors may help advance the development of therapeutic options for RASopathy patients.


Subject(s)
Ectodermal Dysplasia/genetics , Failure to Thrive/genetics , Heart Defects, Congenital/genetics , Mitogen-Activated Protein Kinases/genetics , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Proto-Oncogene Proteins B-raf/genetics , Alleles , Animals , Ectodermal Dysplasia/metabolism , Ectodermal Dysplasia/pathology , Facies , Failure to Thrive/metabolism , Failure to Thrive/pathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Phenotype , Proto-Oncogene Proteins B-raf/metabolism
5.
Hum Mol Genet ; 30(3-4): 213-225, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33517396

ABSTRACT

Primary cilia contain specific proteins to achieve their functions as cellular antennae. Ciliary protein trafficking is mediated by the intraflagellar transport (IFT) machinery containing the IFT-A and IFT-B complexes. Mutations in genes encoding the IFT-A subunits (IFT43, IFT121/WDR35, IFT122, IFT139/TTC21B, IFT140 and IFT144/WDR19) often result in skeletal ciliopathies, including cranioectodermal dysplasia (CED). We here characterized the molecular and cellular defects of CED caused by compound heterozygous mutations in IFT144 [the missense variant IFT144(L710S) and the nonsense variant IFT144(R1103*)]. These two variants were distinct with regard to their interactions with other IFT-A subunits and with the IFT-B complex. When exogenously expressed in IFT144-knockout (KO) cells, IFT144(L710S) as well as IFT144(WT) rescued both moderately compromised ciliogenesis and the abnormal localization of ciliary proteins. As the homozygous IFT144(L710S) mutation was found to cause autosomal recessive retinitis pigmentosa, IFT144(L710S) is likely to be hypomorphic at the cellular level. In striking contrast, the exogenous expression of IFT144(R1103*) in IFT144-KO cells exacerbated the ciliogenesis defects. The expression of IFT144(R1103*) together with IFT144(WT) restored the abnormal phenotypes of IFT144-KO cells. However, the coexpression of IFT144(R1103*) with the hypomorphic IFT144(L710S) variant in IFT144-KO cells, which mimics the genotype of compound heterozygous CED patients, resulted in severe ciliogenesis defects. Taken together, these observations demonstrate that compound heterozygous mutations in IFT144 cause severe ciliary defects via a complicated mechanism, where one allele can cause severe ciliary defects when combined with a hypomorphic allele.


Subject(s)
Bone and Bones/abnormalities , Cilia/metabolism , Craniosynostoses/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Ectodermal Dysplasia/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mutation , Bone and Bones/metabolism , Bone and Bones/physiopathology , Cilia/pathology , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/physiopathology , Codon, Nonsense , Craniosynostoses/genetics , Craniosynostoses/physiopathology , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/physiopathology , HEK293 Cells , Humans , Mutation, Missense
6.
Methods Mol Biol ; 2248: 167-183, 2021.
Article in English | MEDLINE | ID: mdl-33185875

ABSTRACT

Genetic deficiency of ectodysplasin A (EDA) causes X-linked hypohidrotic ectodermal dysplasia, a congenital condition characterized by the absence or abnormal formation of sweat glands, teeth, and several skin appendages. Stimulation of the EDA receptor (EDAR) with agonists in the form of recombinant EDA or anti-EDAR antibodies can compensate for the absence of Eda in a mouse model of Eda deficiency, provided that agonists are administered in a timely manner during fetal development. Here we provide detailed protocols for the administration of EDAR agonists or antagonists, or other proteins, by the intravenous, intraperitoneal, and intra-amniotic routes as well as protocols to collect blood, to visualize sweat gland function, and to prepare skulls in mice.


Subject(s)
Edar Receptor/metabolism , Signal Transduction/drug effects , Animals , Animals, Newborn , Disease Models, Animal , Drug Administration Routes , Ectodermal Dysplasia/drug therapy , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Edar Receptor/genetics , Mice , Phenotype , Recombinant Proteins/administration & dosage , Treatment Outcome
7.
Mech Ageing Dev ; 194: 111411, 2021 03.
Article in English | MEDLINE | ID: mdl-33309600

ABSTRACT

Senescence is a biological process that induces a permanent cell cycle arrest and a specific gene expression program in response to various stressors. Following studies over the last few decades, the concept of senescence has evolved from an antiproliferative mechanism in cancer (oncogene-induced senescence) to a critical component of physiological processes associated with embryonic development, tissue regeneration, ageing and its associated diseases. In somatic cells, oncogenic mutations in RAS-MAPK pathway genes are associated with oncogene-induced senescence and cancer, while germline mutations in the same pathway are linked to a group of monogenic developmental disorders generally termed RASopathies. Here, we consider that in these disorders, senescence induction may result in opposing outcomes, a tumour protective effect and a possible contributor to a premature ageing phenotype identified in Costello syndrome, which belongs to the RASopathy group. In this review, we will highlight the role of senescence in organismal homeostasis and we will describe the current knowledge about senescence in RASopathies. Additionally, we provide a perspective on examples of experimentally characterised RASopathy mutations that, alone or in combination with various stressors, may also trigger an age-dependent chronic senescence, possibly contributing to the age-dependent worsening of RASopathy pathophenotype and the reduction of lifespan.


Subject(s)
Aging, Premature/metabolism , Aging/metabolism , Cell Proliferation , Cellular Senescence , Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism , Age Factors , Aging/genetics , Aging/pathology , Aging, Premature/genetics , Aging, Premature/pathology , Animals , Cell Differentiation , Costello Syndrome/genetics , Costello Syndrome/metabolism , Costello Syndrome/pathology , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Ectodermal Dysplasia/pathology , Facies , Failure to Thrive/genetics , Failure to Thrive/metabolism , Failure to Thrive/pathology , Genetic Predisposition to Disease , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Humans , Mutation , Noonan Syndrome/genetics , Noonan Syndrome/metabolism , Noonan Syndrome/pathology , Phenotype , Signal Transduction , ras Proteins/genetics
8.
Gene ; 733: 144369, 2020 Apr 05.
Article in English | MEDLINE | ID: mdl-31972311

ABSTRACT

MAP2K1 encodes mitogen-activated protein kinase 1 (MEK1). Mutations in MAP2K1 lead to continuous activation of MEK/ERK signaling pathway, giving rise to cardio-facio-cutaneous syndrome (CFCS). However, the molecular mechanisms of abnormal activation of MEK/ERK signaling pathway and the role of autophagy, if any, in manifesting CFCS in MAP2K mutants remain unclear. Here, we report three Chinese children with CFCS having MAP2K1 pathogenic variants, identified by exome sequencing. They presented with dysmorphic facial features, seizures, psychomotor retardation, and short stature. Additionally, the third child showed pulmonary valve stenosis, multiple skeletal deformities, and osteoporosis. Whole exome sequencing revealed two heterozygous missense mutations in exon 3 of MAP2K1 (c.383G>T; p.Gly128Val and c.389A>G; p.Tyr130Cys), as well as a novel heterozygous missense variant (c.170A>T; p.Lys57Met) in exon 2 of MAP2K1. In SH-SY5Y cells, we identified, for the first time, that MAP2K1 mutations can activate the p-ERK-dependent cell cycle progression and autophagy, and cause CFCS. Our results extended the mutational spectrum of MAP2K1, examined the role of MEK1 protein in nerve cell functions, and demonstrated, for the first time, that autophagy may mediate the altered MAP2K1 function, leading to CFCS phenotypes.


Subject(s)
Autophagy , Ectodermal Dysplasia/pathology , Failure to Thrive/pathology , Heart Defects, Congenital/pathology , MAP Kinase Kinase 1/genetics , Mutation , Adult , Apoptosis , Cell Cycle , Cell Movement , Cell Proliferation , Child , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Facies , Failure to Thrive/genetics , Failure to Thrive/metabolism , Female , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Humans , Infant , MAP Kinase Signaling System , Male , Phenotype , Phosphorylation , Tumor Cells, Cultured
9.
Epigenetics Chromatin ; 12(1): 31, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31164150

ABSTRACT

The transcription factor p63 regulates epidermal genes and the enhancer landscape in skin keratinocytes. Its molecular function in controlling the chromatin structure is, however, not yet completely understood. Here, we integrated multi-omics profiles, including the transcriptome, transcription factor DNA-binding and chromatin accessibility, in skin keratinocytes isolated from EEC syndrome patients carrying p63 mutations, to examine the role of p63 in shaping the chromatin architecture. We found decreased chromatin accessibility in p63- and CTCF-bound open chromatin regions that potentially contributed to gene deregulation in mutant keratinocytes. Cooperation of p63 and CTCF seemed to assist chromatin interactions between p63-bound enhancers and gene promoters in skin keratinocytes. Our study suggests an intriguing model where cell type-specific transcription factors such as p63 cooperate with the genome organizer CTCF in the three-dimensional chromatin space to regulate the transcription program important for the proper cell identity.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Keratinocytes/metabolism , Skin/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , CCCTC-Binding Factor/genetics , Cell Differentiation/physiology , Chromatin/genetics , Chromatin Assembly and Disassembly/physiology , Cleft Lip/genetics , Cleft Lip/metabolism , Cleft Palate/genetics , Cleft Palate/metabolism , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Humans , Keratinocytes/cytology , Mutation , Phosphoproteins/metabolism , Primary Cell Culture , Skin/cytology , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor Proteins/genetics
10.
Am J Med Genet A ; 179(8): 1415-1419, 2019 08.
Article in English | MEDLINE | ID: mdl-31119873

ABSTRACT

Incontinentia Pigmenti (IP; MIM 308300) is an X-linked dominant genodermatosis caused by pathogenic variant in IKBKG. The phenotype in adults is poorly described compared to that in children. Questionnaire survey of 99 affected women showed an age at diagnosis from newborn to 41 years, with 53 diagnosed by 6 months of age and 30 as adults. Stage I, II, and III lesions persisted in 16%, 17%, and 71%, respectively, of those who had ever had them. IP is allelic to two forms of ectodermal dysplasia. Many survey respondents reported hypohidrosis and/or heat intolerance and most had Stage IV findings. This suggests that "Stage IV" may be congenitally dysplastic skin that becomes more noticeable with maturity. Fifty-one had dentures or implants with 26 having more invasive jaw or dental surgery. Half had wiry or uncombable hair. Seventy-three reported abnormal nails with 27 having long-term problems. Cataracts and retinal detachment were the reported causes of vision loss. Four had microphthalmia. Respondents without genetic confirmation of IP volunteered information suggesting more involved phenotype or possibly misassigned diagnosis. Ascertainment bias likely accounts for the low prevalence of neurocognitive problems in the respondents.


Subject(s)
Cataract/genetics , Ectodermal Dysplasia/genetics , I-kappa B Kinase/genetics , Incontinentia Pigmenti/genetics , Mutation , Retinal Detachment/genetics , Adolescent , Adult , Aged , Cataract/diagnosis , Cataract/metabolism , Cataract/pathology , Dental Implants , Dentures , Ectodermal Dysplasia/diagnosis , Ectodermal Dysplasia/metabolism , Ectodermal Dysplasia/pathology , Female , Gene Expression , Hair/metabolism , Hair/pathology , Humans , I-kappa B Kinase/deficiency , Incontinentia Pigmenti/diagnosis , Incontinentia Pigmenti/metabolism , Incontinentia Pigmenti/pathology , Middle Aged , Nails/metabolism , Nails/pathology , Phenotype , Retinal Detachment/diagnosis , Retinal Detachment/metabolism , Retinal Detachment/pathology , Severity of Illness Index , Skin/metabolism , Skin/pathology , Surveys and Questionnaires , Tooth/metabolism , Tooth/pathology
11.
Am J Med Genet A ; 179(3): 442-447, 2019 03.
Article in English | MEDLINE | ID: mdl-30703280

ABSTRACT

An international advisory group met at the National Institutes of Health in Bethesda, Maryland in 2017, to discuss a new classification system for the ectodermal dysplasias (EDs) that would integrate both clinical and molecular information. We propose the following, a working definition of the EDs building on previous classification systems and incorporating current approaches to diagnosis: EDs are genetic conditions affecting the development and/or homeostasis of two or more ectodermal derivatives, including hair, teeth, nails, and certain glands. Genetic variations in genes known to be associated with EDs that affect only one derivative of the ectoderm (attenuated phenotype) will be grouped as non-syndromic traits of the causative gene (e.g., non-syndromic hypodontia or missing teeth associated with pathogenic variants of EDA "ectodysplasin"). Information for categorization and cataloging includes the phenotypic features, Online Mendelian Inheritance in Man number, mode of inheritance, genetic alteration, major developmental pathways involved (e.g., EDA, WNT "wingless-type," TP63 "tumor protein p63") or the components of complex molecular structures (e.g., connexins, keratins, cadherins).


Subject(s)
Ectodermal Dysplasia/diagnosis , Ectodermal Dysplasia/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Phenotype , Alleles , Biomarkers , Databases, Genetic , Ectodermal Dysplasia/metabolism , Humans , Signal Transduction
12.
Small GTPases ; 10(3): 210-217, 2019 05.
Article in English | MEDLINE | ID: mdl-28287327

ABSTRACT

DOCK6 is a RAC1/CDC42 guanine nucleotide exchange factor, however, little is known about its function and sub-cellular localization. DOCK6 regulates the balance between RAC1 and RHOA activity during cell adhesion and is important for CDC42-dependent mitotic chromosome alignment. Surprisingly, a cell intrinsic adaptation mechanism compensates for errors in these DOCK6 functions that arise as a consequence of prolonged DOCK6 depletion or complete removal in DOCK6 knockout cells. Down-regulation of the ubiquitin-like modifier ISG15 accounts for this adaptation. Strikingly, although most other DOCK family proteins are deployed on the plasma membrane, here we show that DOCK6 localizes to the endoplasmic reticulum (ER) in dependence of its DHR-1 domain. ER localization of DOCK6 opens up new insights into its functions.


Subject(s)
Cell Membrane/metabolism , Cytokines/metabolism , Endoplasmic Reticulum/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Ubiquitins/metabolism , Cell Membrane/genetics , Cytokines/genetics , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Ectodermal Dysplasia/pathology , Endoplasmic Reticulum/genetics , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/metabolism , Limb Deformities, Congenital/pathology , Scalp Dermatoses/congenital , Scalp Dermatoses/genetics , Scalp Dermatoses/metabolism , Scalp Dermatoses/pathology , Ubiquitins/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
13.
Hum Mol Genet ; 28(1): 74-83, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30239744

ABSTRACT

Cardio-facio-cutaneous (CFC) syndrome, a genetic disorder caused by germline mutations in BRAF, KRAS, MAP2K1 and MAP2K2, is characterized by growth retardation, heart defects, dysmorphic facial appearance and dermatologic abnormalities. We have previously reported that knock-in mice expressing the CFC syndrome-associated mutation, Braf Q241R, showed growth retardation because of gastrointestinal dysfunction. However, other factors associated with growth retardation, including chondrogenesis and endocrinological profile, have not been examined. Here, we show that 3- and 4-week-old BrafQ241R/+ mice have decreased body weight and length, as well as reduced growth plate width in the proximal tibiae. Furthermore, proliferative and hypertrophic chondrocyte zones of the growth plate were reduced in BrafQ241R/+ mice compared with Braf+/+ mice. Immunohistological analysis revealed that extracellular signal-regulated kinase (ERK) activation was enhanced in hypertrophic chondrocytes in BrafQ241R/+ mice. In accordance with growth retardation and reduced growth plate width, decreased serum levels of insulin-like growth factor 1 (IGF-1) and IGF binding protein 3 (IGFBP-3) were observed in BrafQ241R/+ mice at 3 and 4 weeks of age. Treatment with C-type natriuretic peptide (CNP), a stimulator of endochondral bone growth and a potent inhibitor of the FGFR3-RAF1-MEK/ERK signaling, increased body and tail lengths in Braf+/+ and BrafQ241R/+ mice. In conclusion, ERK activation in chondrocytes and low serum IGF-1/IGFBP-3 levels could be associated with the growth retardation observed in BrafQ241R/+ mice. Our data also suggest that CNP is a potential therapeutic target in CFC syndrome.


Subject(s)
Ectodermal Dysplasia/metabolism , Failure to Thrive/metabolism , Heart Defects, Congenital/metabolism , Natriuretic Peptide, C-Type/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Animals , Chondrocytes/physiology , Disease Models, Animal , Ectodermal Dysplasia/physiopathology , Facies , Failure to Thrive/physiopathology , Germ-Line Mutation , Growth Disorders/metabolism , Heart Defects, Congenital/physiopathology , Humans , Insulin-Like Growth Factor I/analysis , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred ICR , Mutation , Natriuretic Peptide, C-Type/metabolism , Proto-Oncogene Proteins B-raf/physiology
14.
J Clin Invest ; 129(2): 583-597, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30422821

ABSTRACT

X-linked dominant incontinentia pigmenti (IP) and X-linked recessive anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) are caused by loss-of-function and hypomorphic IKBKG (also known as NEMO) mutations, respectively. We describe a European mother with mild IP and a Japanese mother without IP, whose 3 boys with EDA-ID died from ID. We identify the same private variant in an intron of IKBKG, IVS4+866 C>T, which was inherited from and occurred de novo in the European mother and Japanese mother, respectively. This mutation creates a new splicing donor site, giving rise to a 44-nucleotide pseudoexon (PE) generating a frameshift. Its leakiness accounts for NF-κB activation being impaired but not abolished in the boys' cells. However, aberrant splicing rates differ between cell types, with WT NEMO mRNA and protein levels ranging from barely detectable in leukocytes to residual amounts in induced pluripotent stem cell-derived (iPSC-derived) macrophages, and higher levels in fibroblasts and iPSC-derived neuronal precursor cells. Finally, SRSF6 binds to the PE, facilitating its inclusion. Moreover, SRSF6 knockdown or CLK inhibition restores WT NEMO expression and function in mutant cells. A recurrent deep intronic splicing mutation in IKBKG underlies a purely quantitative NEMO defect in males that is most severe in leukocytes and can be rescued by the inhibition of SRSF6 or CLK.


Subject(s)
Ectodermal Dysplasia , Frameshift Mutation , I-kappa B Kinase , Incontinentia Pigmenti , Introns , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Ectodermal Dysplasia/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , I-kappa B Kinase/deficiency , I-kappa B Kinase/metabolism , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/pathology , Incontinentia Pigmenti/genetics , Incontinentia Pigmenti/metabolism , Incontinentia Pigmenti/pathology , Macrophages/metabolism , Macrophages/pathology , Male
15.
Nagoya J Med Sci ; 80(3): 299-307, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30214079

ABSTRACT

The Notch signaling pathway is highly conserved and essential for animal development. It is required for cell differentiation, survival, and proliferation. Regulation of Notch signaling is a crucial process for human health. Ligands initiate a signal cascade by binding to Notch receptors expressed on a neighboring cell. Notch receptors interact with ligands through their epidermal growth factor-like repeats (EGF repeats). Most EGF repeats are modified by O-glycosylation with residues such as O-linked N-acetylglucosamine (O-GlcNAc), O-fucose, and O-glucose. These O-glycan modifications are important for Notch function. Defects in O-glycosylation affect Notch-ligand interaction, trafficking of Notch receptors, and Notch stability on the cell surface. Although the roles of each modification are not fully understood, O-fucose is essential for binding of Notch receptors to their ligands. We reported an EGF domain-specific O-GlcNAc transferase (EOGT) localized in the endoplasmic reticulum. Mutations in genes encoding EOGT or NOTCH1 cause Adams-Oliver syndrome. Dysregulation of Notch signaling because of defects or mutations in Notch receptors or Notch signal-regulating proteins, such as glycosyltransferases, induce a variety of congenital disorders. In this review, we discuss O-glycosylation of Notch receptors and congenital human diseases caused by defects in O-glycans on Notch receptors.


Subject(s)
Receptors, Notch/metabolism , Animals , Ectodermal Dysplasia/metabolism , Epidermal Growth Factor/metabolism , Glycosylation , Humans , Limb Deformities, Congenital/metabolism , N-Acetylglucosaminyltransferases/metabolism , Scalp Dermatoses/congenital , Scalp Dermatoses/metabolism
16.
Cell Reprogram ; 20(4): 215-224, 2018 08.
Article in English | MEDLINE | ID: mdl-29989433

ABSTRACT

Ectrodactyly-Ectodermal dysplasia-Clefting (EEC) syndrome is a rare monogenic disease with autosomal dominant inheritance caused by mutations in the TP63 gene, leading to progressive corneal keratinocyte loss, limbal stem cell deficiency (LSCD), and eventually blindness. Currently, there is no treatment available to cure or slow down the keratinocyte loss. Human oral mucosal epithelial stem cells (hOMESCs), which are a mixed population of keratinocyte precursor stem cells, are used as source of autologous tissue for treatment of bilateral LSCD. However, hOMESCs from EEC patients have a reduced life span due to TP63 mutations and cannot be used for autologous transplantation. Human induced pluripotent stem cells (hiPSCs) represent a potentially unlimited source of autologous limbal stem cell for EEC patients and can be genetically modified by genome editing technologies to correct the disease ex vivo before transplantation. In this study, we describe for the first time the generation of integration-free EEC-hiPSCs from hOMESCs of EEC patients by Sendai virus vector and episomal vector-based reprogramming. The generated hiPSC clones expressed pluripotency markers and were successfully differentiated into derivatives of the three germ layers, as well as toward corneal epithelium. These cells may be used for EEC disease modeling and open perspectives for applications in cell therapy of LSCD.


Subject(s)
Biomarkers/analysis , Cell Differentiation , Cleft Lip/pathology , Cleft Palate/pathology , Ectodermal Dysplasia/pathology , Induced Pluripotent Stem Cells/pathology , Mouth Mucosa/pathology , Cells, Cultured , Cleft Lip/genetics , Cleft Lip/metabolism , Cleft Palate/genetics , Cleft Palate/metabolism , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mouth Mucosa/metabolism , Mutation , Phenotype , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
18.
Birth Defects Res ; 110(4): 376-381, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29134781

ABSTRACT

BACKGROUND: Sensenbrenner syndrome (cranioectodermal dysplasia, CED) is a very rare autosomal recessive ciliopathy first described by Judith Sensenbrenner in 1975. CED is a complex disorder characterized by craniofacial, skeletal, and ectodermal abnormalities. The clinical symptoms are variable and the CED phenotype may present intrafamilial and interfamilial differences. Sensenbrenner syndrome belongs to a group of ciliary chondrodysplasias and is a genetically heterogeneous disease. Mutations in six genes: IFT122, WDR35, IFT43, WDR19, IFT52, and IFT140 have been associated with this disorder. All known CED genes encode proteins that are part of the intraflagellar transport complex, which plays an important role in the assembly and maintenance of cilia. CASE: We report a on 2-year-old male patient affected by Sensenbrenner syndrome. Dysmorphic features included short stature with rhizomelic shortening of limbs, short fingers, narrow chest, high forehead, epicanthal folds, telecanthus, broad nasal bridge, low-set ears, sparse hair, and widely space teeth. Craniosynostosis was surgically corrected at the age of 4 months. The patient presented chronic renal disease. Nephrologic picture showed early stages of nephronophthisis. Psychomotor development was apparently normal. Molecular analysis of the affected individual revealed compound heterozygosity for a novel nonsense p.(Arg113*) and a missense p.(Asp841Val) variant in the WDR35 gene. CONCLUSIONS: The observations of the CED patient in this study provide additional clinical data and expand the molecular spectrum of Sensenbrenner syndrome. Moreover, the two variants identified in the proband provide further evidence for the WDR35 mutations as the most common cause of this rare syndrome.


Subject(s)
Bone and Bones/abnormalities , Craniosynostoses/genetics , Ectodermal Dysplasia/genetics , Mutation, Missense , Proteins , Amino Acid Substitution , Bone and Bones/metabolism , Bone and Bones/physiology , Child, Preschool , Craniosynostoses/metabolism , Cytoskeletal Proteins , Ectodermal Dysplasia/metabolism , Hedgehog Proteins , Humans , Intracellular Signaling Peptides and Proteins , Male
19.
Mol Psychiatry ; 23(8): 1687-1698, 2018 08.
Article in English | MEDLINE | ID: mdl-29158583

ABSTRACT

Ras/MAPK pathway signaling is a major participant in neurodevelopment, and evidence suggests that BRAF, a key Ras signal mediator, influences human behavior. We studied the role of the mutation BRAFQ257R, the most common cause of cardiofaciocutaneous syndrome (CFC), in an induced pluripotent stem cell (iPSC)-derived model of human neurodevelopment. In iPSC-derived neuronal cultures from CFC subjects, we observed decreased p-AKT and p-ERK1/2 compared to controls, as well as a depleted neural progenitor pool and rapid neuronal maturation. Pharmacological PI3K/AKT pathway manipulation recapitulated cellular phenotypes in control cells and attenuated them in CFC cells. CFC cultures displayed altered cellular subtype ratios and increased intrinsic excitability. Moreover, in CFC cells, Ras/MAPK pathway activation and morphological abnormalities exhibited cell subtype-specific differences. Our results highlight the importance of exploring specific cellular subtypes and of using iPSC models to reveal relevant human-specific neurodevelopmental events.


Subject(s)
Ectodermal Dysplasia/metabolism , Failure to Thrive/metabolism , Heart Defects, Congenital/metabolism , Induced Pluripotent Stem Cells/metabolism , MAP Kinase Signaling System , Neurogenesis/physiology , Neurons/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Cell Culture Techniques , Ectodermal Dysplasia/pathology , Facies , Failure to Thrive/pathology , Heart Defects, Congenital/pathology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mutation , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Neurons/pathology , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism
20.
Cell Mol Life Sci ; 75(7): 1179-1190, 2018 04.
Article in English | MEDLINE | ID: mdl-29103147

ABSTRACT

The transcription factor p63 is a master regulator of epidermal development. Mutations in p63 give rise to human developmental diseases that often manifest epidermal defects. In this review, we summarize major p63 isoforms identified so far and p63 mutation-associated human diseases that show epidermal defects. We discuss key roles of p63 in epidermal keratinocyte proliferation and differentiation, emphasizing its master regulatory control of the gene expression pattern and epigenetic landscape that define epidermal fate. We subsequently review the essential function of p63 during epidermal commitment and transdifferentiation towards epithelial lineages, highlighting the notion that p63 is the guardian of the epithelial lineage. Finally, we discuss current therapeutic development strategies for p63 mutation-associated diseases. Our review proposes future directions for dissecting p63-controlled mechanisms in normal and diseased epidermal development and for developing therapeutic options.


Subject(s)
Epidermis/metabolism , Gene Expression Profiling , Keratinocytes/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Ectodermal Dysplasia/therapy , Humans , Mutation , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...