Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Biochem Biophys Res Commun ; 598: 74-80, 2022 04 02.
Article in English | MEDLINE | ID: mdl-35151207

ABSTRACT

The histone methyltransferase SET domain bifurcated 1 (SETDB1) catalyzes the trimethylation of lysine 9 of histone H3, thereby regulating gene expression. In this study, we used conditional knockout mice, where Setdb1 was deleted only in neural crest cells (Setdb1fl/fl,Wnt1-Cre + mice), to clarify the role of SETDB1 in palatal development. Setdb1fl/fl,Wnt1-Cre + mice died shortly after birth due to a cleft palate with full penetration. Reduced palatal mesenchyme proliferation was seen in Setdb1fl/fl,Wnt1-Cre + mice, which might be a possible mechanism of cleft palate development. Quantitative RT-PCR and in situ hybridization showed that expression of the Pax9, Bmp4, Bmpr1a, Wnt5a, and Fgf10 genes, known to be important for palatal development, were markedly decreased in the palatal mesenchyme of Setdb1fl/fl,Wnt1-Cre + mice. Along with these phenomena, SMAD1/5/9 phosphorylation was decreased by the loss of Setdb1. Our results demonstrated that SETDB1 is indispensable for palatal development partially through its proliferative effect. Taken together with previous reports that PAX9 regulates BMP signaling during palatal development which implies that loss of Setdb1 may be involved in the cleft palate development by decreasing SMAD-dependent BMP signaling through Pax9.


Subject(s)
Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/physiology , Palate/embryology , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Proliferation/genetics , Cleft Palate/genetics , Histone-Lysine N-Methyltransferase/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Crest/physiopathology , PAX9 Transcription Factor/genetics , PAX9 Transcription Factor/metabolism , Palate/abnormalities , Palate/pathology , Smad Proteins/genetics , Smad Proteins/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
2.
Dev Biol ; 475: 222-233, 2021 07.
Article in English | MEDLINE | ID: mdl-33577830

ABSTRACT

Cells display distinct metabolic characteristics depending on its differentiation stage. The fuel type of the cells serves not only as a source of energy but also as a driver of differentiation. Glucose, the primary nutrient to the cells, is a critical regulator of rapidly growing embryos. This metabolic change is a consequence as well as a cause of changes in genetic program. Disturbance of fetal glucose metabolism such as in diabetic pregnancy is associated with congenital heart disease. In utero hyperglycemia impacts the left-right axis establishment, migration of cardiac neural crest cells, conotruncal formation and mesenchymal formation of the cardiac cushion during early embryogenesis and causes cardiac hypertrophy in late fetal stages. In this review, we focus on the role of glucose in cardiogenesis and the molecular mechanisms underlying heart diseases associated with hyperglycemia.


Subject(s)
Glucose/metabolism , Heart/embryology , Hyperglycemia/physiopathology , Animals , Cell Differentiation/genetics , Female , Heart/physiopathology , Heart Defects, Congenital/genetics , Humans , Neural Crest/physiopathology , Organogenesis/genetics , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology
3.
Biomolecules ; 11(1)2020 12 22.
Article in English | MEDLINE | ID: mdl-33375093

ABSTRACT

The neural crest hypothesis states that the phenotypic features of the domestication syndrome are due to a reduced number or disruption of neural crest cells (NCCs) migration, as these cells differentiate at their final destinations and proliferate into different tissues whose activity is reduced by domestication. Comparing the phenotypic characteristics of modern and prehistoric man, it is clear that during their recent evolutionary past, humans also went through a process of self-domestication with a simultaneous prolongation of the period of socialization. This has led to the development of social abilities and skills, especially language, as well as neoteny. Disorders of neural crest cell development and migration lead to many different conditions such as Waardenburg syndrome, Hirschsprung disease, fetal alcohol syndrome, DiGeorge and Treacher-Collins syndrome, for which the mechanisms are already relatively well-known. However, for others, such as Williams-Beuren syndrome and schizophrenia that have the characteristics of hyperdomestication, and autism spectrum disorders, and 7dupASD syndrome that have the characteristics of hypodomestication, much less is known. Thus, deciphering the biological determinants of disordered self-domestication has great potential for elucidating the normal and disturbed ontogenesis of humans, as well as for the understanding of evolution of mammals in general.


Subject(s)
Biological Evolution , Domestication , Language , Neural Crest/physiopathology , Cell Movement/genetics , Cell Proliferation/genetics , Fetal Alcohol Spectrum Disorders/genetics , Hirschsprung Disease/genetics , Humans , Mandibulofacial Dysostosis/genetics , Neural Crest/growth & development , Neural Crest/metabolism , Phenotype , Schizophrenia/genetics , Social Skills , Waardenburg Syndrome/genetics , Williams Syndrome/genetics
4.
Rev. medica electron ; 42(4): 2049-2065,
Article in Spanish | LILACS, CUMED | ID: biblio-1139295

ABSTRACT

RESUMEN Las células de la cresta neural son pluripotenciales y son llamadas la cuarta hoja germinativa del embrión. Con el objetivo de estructurar los referentes teóricos actualizados que sustenten la afirmación precedente y que constituirá material de estudio para los estudiantes de las Ciencias Médicas, se realizó la revisión de 28 referencias bibliográficas, de ellas 89% actualizadas. Estas células aparecen durante la neurulación y pasado este proceso transitan de epitelial a mesenquimatosa; migran siguiendo señales de la matriz extracelular a todo el cuerpo del embrión diferenciándose en tejidos disimiles. Muy vinculados en su evolución a mecanismos epigenéticos, hacen a esta población celular vulnerables a ser dañadas invocándose en la etiología de diferentes defectos congénitos y enfermedades crónicas no trasmisibles como cáncer. Como conclusión por su pluripotencialidad y por los mecanismos moleculares que distinguen su evolución son consideradas por muchos autores la cuarta hoja germinativa del embrión (AU).


SUMMARY Neural crest cells are pluripotentials, and are called the fourth germinative leaf of the embryo. With the objective of structuring the updated theoretical referents backing up the precedent affirmation that will be study material for the students of Medical Sciences, the authors reviewed 28 bibliographic references, 89 % of them updated. These cells appear during neurulation and after this process they transit from epithelial to mesenchymal; following extracellular matrix signals, they migrate to the whole embryo body differentiating themselves in dissimilar tissues. Tightly related in their evolution to epigenetic mechanisms, this cell population is very likely to be damaged and so they are invoked in the etiology of different congenital defects and noncommunicable chronic diseases like cancer. In conclusion, due to their pluripotentiality and the molecular mechanisms distinguishing their evolution, many authors consider them the embryo´s fourth germinative leaf (AU).


Subject(s)
Humans , Male , Female , Cells/metabolism , Neural Crest/pathology , Students, Medical , Vertebrates/genetics , Neurulation/physiology , Neural Crest/abnormalities , Neural Crest/physiology , Neural Crest/physiopathology
5.
Cell Death Dis ; 10(11): 812, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31649239

ABSTRACT

Head dysgenesis is a major cause of fetal demise and craniofacial malformation. Although mutations in genes of the head ontogenetic program have been reported, many cases remain unexplained. Head dysgenesis has also been related to trisomy or amplification of the chromosomal region overlapping the CDX2 homeobox gene, a master element of the trunk ontogenetic program. Hence, we investigated the repercussion on head morphogenesis of the imbalance between the head and trunk ontogenetic programs, by means of ectopic rostral expression of CDX2 at gastrulation. This caused severe malformations affecting the forebrain and optic structures, and also the frontonasal process associated with defects in neural crest cells colonization. These malformations are the result of the downregulation of genes of the head program together with the abnormal induction of trunk program genes. Together, these data indicate that the imbalance between the anterior and posterior ontogenetic programs in embryos is a new possible cause of head dysgenesis during human development, linked to defects in setting up anterior neuroectodermal structures.


Subject(s)
CDX2 Transcription Factor/genetics , Craniofacial Abnormalities/genetics , Head/physiopathology , Morphogenesis/genetics , Animals , Craniofacial Abnormalities/physiopathology , Embryonic Development/genetics , Gastrulation/genetics , Gene Expression Regulation, Developmental/genetics , Genes, Homeobox/genetics , Head/growth & development , Humans , Mice , Neural Crest/growth & development , Neural Crest/physiopathology , Prosencephalon/growth & development , Prosencephalon/pathology
6.
Dev Biol ; 444 Suppl 1: S110-S143, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29802835

ABSTRACT

The neural crest (NC) is a transient, multipotent and migratory cell population that generates an astonishingly diverse array of cell types during vertebrate development. These cells, which originate from the ectoderm in a region lateral to the neural plate in the neural fold, give rise to neurons, glia, melanocytes, chondrocytes, smooth muscle cells, odontoblasts and neuroendocrine cells, among others. Neurocristopathies (NCP) are a class of pathologies occurring in vertebrates, especially in humans that result from the abnormal specification, migration, differentiation or death of neural crest cells during embryonic development. Various pigment, skin, thyroid and hearing disorders, craniofacial and heart abnormalities, malfunctions of the digestive tract and tumors can also be considered as neurocristopathies. In this review we revisit the current classification and propose a new way to classify NCP based on the embryonic origin of the affected tissues, on recent findings regarding the molecular mechanisms that drive NC formation, and on the increased complexity of current molecular embryology techniques.


Subject(s)
Embryonic Development/physiology , Neural Crest/embryology , Neural Crest/physiopathology , Animals , Body Patterning/physiology , Cell Differentiation , Cell Lineage , Cell Movement , Ectoderm , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Humans , Melanocytes/cytology , Neural Crest/cytology , Peripheral Nervous System/embryology , Vertebrates/embryology
7.
Proc Natl Acad Sci U S A ; 114(43): E9046-E9055, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073101

ABSTRACT

Kabuki syndrome, a congenital craniofacial disorder, manifests from mutations in an X-linked histone H3 lysine 27 demethylase (UTX/KDM6A) or a H3 lysine 4 methylase (KMT2D). However, the cellular and molecular etiology of histone-modifying enzymes in craniofacial disorders is unknown. We now establish Kabuki syndrome as a neurocristopathy, whereby the majority of clinical features are modeled in mice carrying neural crest (NC) deletion of UTX, including craniofacial dysmorphism, cardiac defects, and postnatal growth retardation. Female UTX NC knockout (FKO) demonstrates enhanced phenotypic severity over males (MKOs), due to partial redundancy with UTY, a Y-chromosome demethylase-dead homolog. Thus, NC cells may require demethylase-independent UTX activity. Consistently, Kabuki causative point mutations upstream of the JmjC domain do not disrupt UTX demethylation. We have isolated primary NC cells at a phenocritical postmigratory timepoint in both FKO and MKO mice, and genome-wide expression and histone profiling have revealed UTX molecular function in establishing appropriate chromatin structure to regulate crucial NC stem-cell signaling pathways. However, the majority of UTX regulated genes do not experience aberrations in H3K27me3 or H3K4me3, implicating alternative roles for UTX in transcriptional control. These findings are substantiated through demethylase-dead knockin mutation of UTX, which supports appropriate facial development.


Subject(s)
Abnormalities, Multiple/etiology , Face/abnormalities , Hematologic Diseases/etiology , Histone Demethylases/metabolism , Neural Crest/physiopathology , Vestibular Diseases/etiology , Animals , Cell Survival/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Histone Demethylases/genetics , Humans , Lysine/metabolism , Male , Mice, Knockout , Mice, Transgenic , Mutation , Neural Crest/metabolism , Nuclear Proteins/genetics , Skull/embryology
8.
Am J Med Genet A ; 173(6): 1705-1709, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28422456

ABSTRACT

We evaluated a family with three siblings, two of whom ages 2 years and 19 months, had long segment colonic agangliosis and anisocoria. The mother also had anisocoria. All three affected family members were mildly dysmorphic with a flat facial profile, square appearance to the face, depressed nasal bridge, and anteverted nares. Genetic testing identified a novel heterozygous mutation, c.234C>G, resulting in a premature stop codon in exon 1 of the PHOX2B gene. Screening for neural crest tumors was performed in the siblings and to date has been negative. This family supports a strong association between non polyalanine tract mutations, autonomic dysfunction, and Hirschsprung disease, but suggests mutation outside of the polyalanine tract may not dictate severe phenotype with significant respiratory compromise. A unique finding in this family is the association of congenital heart disease in two of the affected patients. These malformations may be a sporadic isolated finding or the result of environmental factors or a modifying allele. Given the association between congenital heart disease and aberrant neural crest cell development, however, findings are suggestive that congenital heart disease may be a rare feature of PHOX2B mutation which has not been previously reported.


Subject(s)
Anisocoria/genetics , Hirschsprung Disease/genetics , Homeodomain Proteins/genetics , Hypoventilation/congenital , Sleep Apnea, Central/genetics , Transcription Factors/genetics , Adult , Anisocoria/physiopathology , Exons/genetics , Face , Female , Heterozygote , Hirschsprung Disease/physiopathology , Humans , Hypoventilation/genetics , Hypoventilation/physiopathology , Infant , Male , Mutation , Neural Crest/growth & development , Neural Crest/physiopathology , Pedigree , Phenotype , Siblings , Sleep Apnea, Central/physiopathology
9.
Gastroenterology ; 153(1): 139-153.e8, 2017 07.
Article in English | MEDLINE | ID: mdl-28342760

ABSTRACT

BACKGROUND & AIMS: Hirschsprung disease is caused by failure of enteric neural crest cells (ENCCs) to fully colonize the bowel, leading to bowel obstruction and megacolon. Heterozygous mutations in the coding region of the RET gene cause a severe form of Hirschsprung disease (total colonic aganglionosis). However, 80% of HSCR patients have short-segment Hirschsprung disease (S-HSCR), which has not been associated with genetic factors. We sought to identify mutations associated with S-HSCR, and used the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing system to determine how mutations affect ENCC function. METHODS: We created induced pluripotent stem cell (iPSC) lines from 1 patient with total colonic aganglionosis (with the G731del mutation in RET) and from 2 patients with S-HSCR (without a RET mutation), as well as RET+/- and RET-/- iPSCs. IMR90-iPSC cells were used as the control cell line. Migration and differentiation capacities of iPSC-derived ENCCs were analyzed in differentiation and migration assays. We searched for mutation(s) associated with S-HSCR by combining genetic and transcriptome data from patient blood- and iPSC-derived ENCCs, respectively. Mutations in the iPSCs were corrected using the CRISPR/Cas9 system. RESULTS: ENCCs derived from all iPSC lines, but not control iPSCs, had defects in migration and neuronal lineage differentiation. RET mutations were associated with differentiation and migration defects of ENCCs in vitro. Genetic and transcriptome analyses associated a mutation in the vinculin gene (VCL M209L) with S-HSCR. CRISPR/Cas9 correction of the RET G731del and VCL M209L mutations in iPSCs restored the differentiation and migration capacities of ENCCs. CONCLUSIONS: We identified mutations in VCL associated with S-HSCR. Correction of this mutation in iPSC using CRISPR/Cas9 editing, as well as the RET G731del mutation that causes Hirschsprung disease with total colonic aganglionosis, restored ENCC function. Our study demonstrates how human iPSCs can be used to identify disease-associated mutations and determine how they affect cell functions and contribute to pathogenesis.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing/methods , Hirschsprung Disease/genetics , Neural Crest/physiopathology , Proto-Oncogene Proteins c-ret/genetics , Vinculin/genetics , Cell Differentiation/genetics , Cell Line , Cell Movement/genetics , DNA Mutational Analysis/methods , Humans , Induced Pluripotent Stem Cells/physiology , Phenotype
10.
Pediatr Surg Int ; 32(12): 1095-1101, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27663687

ABSTRACT

PURPOSE: Hirschsprung's disease (HD) is caused by a failure of enteric neural crest-derived cells (ENCC) to colonize the bowel, resulting in an absence of the enteric nervous system (ENS). Previously, we developed a Sox10 transgenic version of the Endothelin receptor-B (Ednrb) mouse to visualize ENCC with the green fluorescent protein, Venus. The aim of this study was to isolate Sox10-Venus+ cells, which are differentiated neurons and glial cells in the ENS, and analyze these cells using Sox10-Venus mice gut. METHODS: The mid-and hindgut of Sox10-Venus+/Ednrb +/+ and Sox10-Venus+/Ednrb -/- at E13.5 and E15.5 were dissected and cells were dissociated. Sox10-Venus+ cells were then isolated. Expression of PGP9.5 and GFAP were evaluated neurospheres using laser scanning microscopy. RESULTS: 7 days after incubation, Sox10-Venus+ cells colonized the neurosphere. There were no significant differences in PGP9.5 expressions on E13.5 and E15.5. GFAP was significantly increased in HD compared to controls on E15.5 (P < 0.05). CONCLUSIONS: Our results suggest increased glial differentiation causes an imbalance in ENCC lineages, leading to a disruption of normal ENS development in this HD model. Isolation of ENCC provides an opportunity to investigate the ENS with purity and might be a useful tool for modeling cell therapy approaches to HD.


Subject(s)
Cell Differentiation/physiology , Enteric Nervous System/embryology , Hirschsprung Disease/embryology , Neural Crest/embryology , Receptor, Endothelin B/physiology , Animals , Cell Culture Techniques , Disease Models, Animal , Enteric Nervous System/physiopathology , Fluorescent Antibody Technique , Intestines/embryology , Intestines/physiopathology , Mice , Mice, Knockout , Neural Crest/physiopathology , Neurons/physiology
11.
Dev Biol ; 417(2): 209-16, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27321561

ABSTRACT

Hirschsprung disease (HSCR, OMIM 142623) is a neurocristopathy caused by a failure of the enteric nervous system (ENS) progenitors derived from neural crest cells (NCCs), to migrate, proliferate, differentiate or survive to and within the gastrointestinal tract, resulting in aganglionosis in the distal colon. The formation of the ENS is a complex process, which is regulated by a large range of molecules and signalling pathways involving both the NCCs and the intestinal environment. This tightly regulated process needs correct regulation of the expression of ENS specific genes. Alterations in the expression of these genes can have dramatic consequences. Several mechanisms that control the expression of genes have been described, such as DNA modification (epigenetic mechanisms), regulation of transcription (transcription factor, enhancers, repressors and silencers), post-transcriptional regulation (3'UTR and miRNAs) and regulation of translation. In this review, we focus on the epigenetic DNA modifications that have been described so far in the context of the ENS development. Moreover we describe the changes that are found in relation to the onset of HSCR.


Subject(s)
Enteric Nervous System/embryology , Gastrointestinal Tract/innervation , Hirschsprung Disease/embryology , Hirschsprung Disease/pathology , Neural Crest/physiopathology , Organogenesis/physiology , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Gastrointestinal Tract/embryology , Hirschsprung Disease/genetics , Histones/metabolism , Humans , Neural Crest/cytology , Organogenesis/genetics , RNA Processing, Post-Transcriptional/genetics , Signal Transduction
12.
Pediatr Surg Int ; 32(1): 3-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26546339

ABSTRACT

BACKGROUND/AIM: The behavior of enteric neural crest-derived cells (ENCC) during enteric nervous system (ENS) development is being gradually understood with the introduction of live-cell imaging. However, many of the analyses to date are two-dimensional and the precise multidirectional migration of ENCC has been challenging to interpret. Mice lacking the endothelin-B receptor gene, Ednrb (-/-) mice, are widely used as a model for Hirschsprung's disease (HD). We have recently developed a Sox10 transgenic (Tg) mouse to visualize ENCC with enhanced green fluorescent protein (Venus). By breeding these two models, we have created a Venus-positive, Sox10 Tg mouse with a deletion of the Ednrb gene, Sox10-Venus(+)/Ednrb (-/-) mouse, to investigate the ENS in HD. The aim of this study was to investigate the behavior of migrating ENCC in the hindgut of the Sox10-Venus(+)/Ednrb (-/-) mouse using three-dimensional and four-dimensional image analysis software. METHODS: To compare the ENCC behavior when the wavefront of ENCC reaches the mid-hindgut between HD mouse and control, we harvested the fetal hindguts of Sox10-Venus(+)/Ednrb (-/-) mice on embryonic day 15.5 (E15.5) and Sox10-Venus(+)/Ednrb (+/+) mice on E12.5, which was used as control. Dissected hindguts were cultured for 360 min and the time-lapse images were obtained using a confocal laser-scanning microscope. Each ENCC at the wavefront was tracked after adjusting the longitudinal axis of the gut to the Y axis and analyzed using Imaris software. RESULTS: Track displacement (TD)-Y indicates ENCC advancement in a rostral-caudal direction. TD-X and TD-Z indicate ENCC advancement perpendicular to the rostral-caudal axis. Mean TD-Y was 34.56 µm in HD, but 63.48 µm in controls. TD-Y/TD-XZ was not significantly different in both groups. However, the mean track speeds were significantly decreased in HD (72.87 µm/h) compared to controls (248.29 µm/h). CONCLUSIONS: Our results showed that the track speed of ENCC advancement was markedly decreased in the HD mice compared to controls. This technique provides added information by tracking ENCC with depth perception, which has potential for further elucidating the altered behavior of ENCC in HD.


Subject(s)
Enteric Nervous System/physiopathology , Hirschsprung Disease/physiopathology , Imaging, Three-Dimensional/methods , Neural Crest/physiopathology , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Disease Models, Animal , Mice , Mice, Transgenic , Microscopy, Confocal , Signal Transduction/physiology
13.
Exp Cell Res ; 325(2): 138-47, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24509233

ABSTRACT

Neural crest cells appear early during embryogenesis and give rise to many structures in the mature adult. In particular, a specific population of neural crest cells migrates to and populates developing cranial tissues. The ensuing differentiation of these cells via individual complex and often intersecting signaling pathways is indispensible to growth and development of the craniofacial complex. Much research has been devoted to this area of development with particular emphasis on cell signaling events required for physiologic development. Understanding such mechanisms will allow researchers to investigate ways in which they can be exploited in order to treat a multitude of diseases affecting the craniofacial complex. Knowing how these multipotent cells are driven towards distinct fates could, in due course, allow patients to receive regenerative therapies for tissues lost to a variety of pathologies. In order to realize this goal, nucleotide sequencing advances allowing snapshots of entire genomes and exomes are being utilized to identify molecular entities associated with disease states. Once identified, these entities can be validated for biological significance with other methods. A crucial next step is the integration of knowledge gleaned from observations in disease states with normal physiology to generate an explanatory model for craniofacial development. This review seeks to provide a current view of the landscape on cell signaling and fate determination of the neural crest and to provide possible avenues of approach for future research.


Subject(s)
Bone Development , Neural Crest/embryology , Neural Crest/physiopathology , Signal Transduction , Skull/embryology , Skull/physiopathology , Animals , Embryonic Development , Humans
14.
J Child Neurol ; 28(6): 768-73, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23576410

ABSTRACT

Neuroblastoma is, at once, the most common and deadly extracranial solid tumor of childhood. Efforts aimed at targeting the neural characteristics of these tumors have taught us much about neural crest cell biology, apoptosis induction in the nervous system, and neurotrophin receptor signaling and intracellular processing. But neuroblastoma remains a formidable enemy to the oncologist and an enigmatic target to the neuroscientist.


Subject(s)
Antineoplastic Agents/administration & dosage , Molecular Targeted Therapy/methods , Neuroblastoma/drug therapy , Neuroblastoma/physiopathology , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Child , Drug Discovery/methods , Humans , Neural Crest/drug effects , Neural Crest/pathology , Neural Crest/physiopathology , Neuroblastoma/genetics , Neuroblastoma/pathology , Precision Medicine/methods , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
15.
J Neurochem ; 124(2): 241-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23134367

ABSTRACT

The neural crest is a unique structure in vertebrates. Wnt1-cre and Wnt1-GAL4 double transgenic (dTg) mice have been used in a variety of studies concerning neural crest cell lineages in which the Cre/loxP or GAL4/UAS system was applied. Here, we show psychiatric disorder-related behavioral abnormalities and histologic alterations in a neural crest-derived brain region in dTg mice. The dTg mice exhibited increased locomotor activity, decreased social interaction, and impaired short-term spatial memory and nesting behavior. The choline acetyltransferase- and vesicular glutamate transporter 2-immunoreactive habenulointerpeduncular fiber tracts that project from the medial habenular nucleus of the epithalamus to the interpeduncular nucleus of the midbrain tegmentum appeared irregular in the dTg mice. Both the medial habenula nucleus and the interpeduncular nucleus were confirmed to be derived from the neural crest. The findings of this study suggest that neural crest-derived cells have pathogenic roles in the development of psychiatric disorders and that the dTg mouse could be a useful animal model for studying the pathophysiology of mental illness such as autism and schizophrenia. Scientists that use the dTg mice as a cre-transgenic deleter line should be cautious in its possible toxicity, especially if behavioral analyses are to be performed.


Subject(s)
Behavior, Animal , Habenula/physiopathology , Mental Disorders/genetics , Neural Crest/abnormalities , Schizophrenia/genetics , Tegmentum Mesencephali/physiopathology , Wnt1 Protein/genetics , Animals , Behavior, Animal/physiology , DNA-Binding Proteins/genetics , Disease Models, Animal , Female , Male , Maze Learning/physiology , Mental Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nesting Behavior/physiology , Neural Crest/physiopathology , Neural Pathways/physiopathology , Saccharomyces cerevisiae Proteins/genetics , Schizophrenia/metabolism , Transcription Factors/genetics , Wnt1 Protein/metabolism
16.
Congenit Anom (Kyoto) ; 51(1): 12-5, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21338411

ABSTRACT

In this review, our work on CHARGE syndrome will be used to exemplify the role of rare cases in birth defects research. The analysis of 29 cases with mutations of CHD7, the causative gene for CHARGE syndrome, clarified the relative importance of the cardinal features, including facial nerve palsy and facial asymmetry. Concurrently, in situ hybridization using chick embryos studies were performed to delineate the expression pattern of Chd7. The Chd7-positive regions in the chick embryos and the anatomical defects commonly seen in patients with CHARGE syndrome were well correlated: expression in the optic placode corresponded with defects such as coloboma, neural tube with mental retardation, and otic placode with ear abnormalities. The correlation between expression in the branchial arches and nasal placode with the clinical symptoms of CHARGE syndrome, however, became apparent when we encountered two unique CHARGE syndrome patients: one with a DiGeorge syndrome phenotype and the other with a Kallman syndrome phenotype. A unifying hypothesis that could explain both the DiGeorge syndrome phenotype and the Kallman syndrome phenotype in patients with CHARGE syndrome may be that the mutation in CHD7 is likely to exert its effect in the common branch of the two pathways of neural crest cells. As exemplified in CHARGE syndrome research, rare cases play a critical role in deciphering the mechanisms of human development. Close collaboration among animal researchers, epidemiologists and clinicians hopefully will enhance and maximize the scientific value of rare cases.


Subject(s)
CHARGE Syndrome/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Animals , CHARGE Syndrome/classification , CHARGE Syndrome/embryology , Chick Embryo , DNA Helicases/biosynthesis , DNA-Binding Proteins/biosynthesis , Female , Humans , In Situ Hybridization , Male , Maternal Exposure , Methimazole/adverse effects , Neural Crest/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects
17.
Am J Med Genet A ; 155A(2): 270-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21271641

ABSTRACT

The craniofacial region is assembled through the active migration of cells and the rearrangement and sculpting of facial prominences and pharyngeal arches, which consequently make it particularly susceptible to a large number of birth defects. Genetic, molecular, and cellular processes must be temporally and spatially regulated to culminate in the three-dimension structures of the face. The starting constituent for the majority of skeletal and connective tissues in the face is a pluripotent population of cells, the cranial neural crest cells (NCCs). In this review we discuss the newest scientific findings in the development of the craniofacial complex as related to NCCs. Furthermore, we present recent findings on NCC diseases called neurocristopathies and, in doing so, provide clinicians with new tools for understanding a growing number of craniofacial genetic disorders.


Subject(s)
Body Patterning , Cell Movement/physiology , Craniofacial Abnormalities/genetics , Neural Crest/embryology , Neural Crest/physiopathology , Signal Transduction/physiology , Skull/embryology , Cell Differentiation/physiology , Cell Proliferation , Craniofacial Abnormalities/pathology , Humans
18.
J Clin Invest ; 121(1): 422-30, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21157040

ABSTRACT

Congenital anomalies of the aortic valve are common and are associated with progressive valvular insufficiency and/or stenosis. In addition, aneurysm, coarctation, and dissection of the ascending aorta and aortic arch are often associated conditions that complicate patient management and increase morbidity and mortality. These associated aortopathies are commonly attributed to turbulent hemodynamic flow through the malformed valve leading to focal defects in the vessel wall. However, numerous surgical and pathological studies have identified widespread cystic medial necrosis and smooth muscle apoptosis throughout the aortic arch in affected patients. Here, we provide experimental evidence for an alternative model to explain the association of aortic vessel and valvular disease. Using mice with primary and secondary cardiac neural crest deficiencies, we have shown that neural crest contribution to the outflow endocardial cushions (the precursors of the semilunar valves) is required for late gestation valvular remodeling, mesenchymal apoptosis, and proper valve architecture. Neural crest was also shown to contribute to the smooth muscle layer of the wall of the ascending aorta and aortic arch. Hence, defects of cardiac neural crest can result in functionally abnormal semilunar valves and concomitant aortic arch artery abnormalities.


Subject(s)
Aortic Valve/embryology , Neural Crest/embryology , Animals , Aortic Valve/abnormalities , Aortic Valve/physiopathology , Apoptosis , Endocardial Cushion Defects/embryology , Endocardial Cushion Defects/physiopathology , Female , Humans , Mice , Mice, Mutant Strains , Models, Cardiovascular , Neural Crest/abnormalities , Neural Crest/physiopathology , PAX3 Transcription Factor , Paired Box Transcription Factors/deficiency , Paired Box Transcription Factors/genetics , Pregnancy , Receptors, Notch/genetics , Receptors, Notch/physiology , Signal Transduction
20.
J Neuroophthalmol ; 28(3): 178-81, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18769279

ABSTRACT

We report a patient with Horner syndrome together with anhidrosis affecting the ipsilateral face and extremities confirmed with starch-iodine and sympathetic skin response testing. No anatomic lesion was apparent. This is the first reported case in which Horner syndrome has been associated with such extensive hemibody sympathetic dysfunction in the absence of other neurologic findings. We propose a developmental disorder of neural crest migration as the cause.


Subject(s)
Autonomic Nervous System Diseases/physiopathology , Horner Syndrome/physiopathology , Hypohidrosis/physiopathology , Sympathetic Nervous System/physiopathology , Adult , Autonomic Nervous System Diseases/diagnosis , Autonomic Nervous System Diseases/etiology , Extremities/innervation , Extremities/physiopathology , Face/blood supply , Face/physiopathology , Functional Laterality/physiology , Galvanic Skin Response , Horner Syndrome/etiology , Humans , Hypohidrosis/etiology , Male , Neural Conduction , Neural Crest/abnormalities , Neural Crest/physiopathology , Neurologic Examination , Pain Measurement , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/physiopathology , Sympathetic Nervous System/abnormalities
SELECTION OF CITATIONS
SEARCH DETAIL
...