Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
PLoS Genet ; 15(3): e1007605, 2019 03.
Article in English | MEDLINE | ID: mdl-30856165

ABSTRACT

Typical Martsolf syndrome is characterized by congenital cataracts, postnatal microcephaly, developmental delay, hypotonia, short stature and biallelic hypomorphic mutations in either RAB3GAP1 or RAB3GAP2. Genetic analysis of 85 unrelated "mutation negative" probands with Martsolf or Martsolf-like syndromes identified two individuals with different homozygous null mutations in ITPA, the gene encoding inosine triphosphate pyrophosphatase (ITPase). Both probands were from multiplex families with a consistent, lethal and highly distinctive disorder; a Martsolf-like syndrome with infantile-onset dilated cardiomyopathy. Severe ITPase-deficiency has been previously reported with infantile epileptic encephalopathy (MIM 616647). ITPase acts to prevent incorporation of inosine bases (rI/dI) into RNA and DNA. In Itpa-null cells dI was undetectable in genomic DNA. dI could be identified at a low level in mtDNA without detectable mitochondrial genome instability, mtDNA depletion or biochemical dysfunction of the mitochondria. rI accumulation was detectable in proband-derived lymphoblastoid RNA. In Itpa-null mouse embryos rI was detectable in the brain and kidney with the highest level seen in the embryonic heart (rI at 1 in 385 bases). Transcriptome and proteome analysis in mutant cells revealed no major differences with controls. The rate of transcription and the total amount of cellular RNA also appeared normal. rI accumulation in RNA-and by implication rI production-correlates with the severity of organ dysfunction in ITPase deficiency but the basis of the cellulopathy remains cryptic. While we cannot exclude cumulative minor effects, there are no major anomalies in the production, processing, stability and/or translation of mRNA.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Cardiomyopathy, Dilated/genetics , Cataract/enzymology , Cataract/genetics , Hypogonadism/enzymology , Hypogonadism/genetics , Intellectual Disability/enzymology , Intellectual Disability/genetics , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/genetics , Pyrophosphatases/deficiency , Animals , Base Sequence , Child, Preschool , DNA Mutational Analysis , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Female , Homozygote , Humans , Inosine/metabolism , Male , Mice , Mice, Knockout , Mouse Embryonic Stem Cells/enzymology , Mutation , Pedigree , Pyrophosphatases/genetics , RNA/genetics , RNA/metabolism , Exome Sequencing
3.
PLoS Genet ; 10(4): e1004242, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24763282

ABSTRACT

Folate-sensitive fragile sites (FSFS) are a rare cytogenetically visible subset of dynamic mutations. Of the eight molecularly characterized FSFS, four are associated with intellectual disability (ID). Cytogenetic expression results from CGG tri-nucleotide-repeat expansion mutation associated with local CpG hypermethylation and transcriptional silencing. The best studied is the FRAXA site in the FMR1 gene, where large expansions cause fragile X syndrome, the most common inherited ID syndrome. Here we studied three families with FRA2A expression at 2q11 associated with a wide spectrum of neurodevelopmental phenotypes. We identified a polymorphic CGG repeat in a conserved, brain-active alternative promoter of the AFF3 gene, an autosomal homolog of the X-linked AFF2/FMR2 gene: Expansion of the AFF2 CGG repeat causes FRAXE ID. We found that FRA2A-expressing individuals have mosaic expansions of the AFF3 CGG repeat in the range of several hundred repeat units. Moreover, bisulfite sequencing and pyrosequencing both suggest AFF3 promoter hypermethylation. cSNP-analysis demonstrates monoallelic expression of the AFF3 gene in FRA2A carriers thus predicting that FRA2A expression results in functional haploinsufficiency for AFF3 at least in a subset of tissues. By whole-mount in situ hybridization the mouse AFF3 ortholog shows strong regional expression in the developing brain, somites and limb buds in 9.5-12.5dpc mouse embryos. Our data suggest that there may be an association between FRA2A and a delay in the acquisition of motor and language skills in the families studied here. However, additional cases are required to firmly establish a causal relationship.


Subject(s)
Fos-Related Antigen-2/genetics , Nuclear Proteins/genetics , Trinucleotide Repeat Expansion/genetics , Alleles , Chromosome Fragile Sites/genetics , DNA Methylation/genetics , Female , Gene Expression/genetics , Humans , Intellectual Disability/genetics , Male , Phenotype , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics
4.
Hum Mol Genet ; 22(16): 3269-82, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23595884

ABSTRACT

Glucocorticoids are vital for the structural and functional maturation of foetal organs, yet excessive foetal exposure is detrimental to adult cardiovascular health. To elucidate the role of glucocorticoid signalling in late-gestation cardiovascular maturation, we have generated mice with conditional disruption of glucocorticoid receptor (GR) in cardiomyocytes and vascular smooth muscle cells using smooth muscle protein 22-driven Cre recombinase (SMGRKO mice) and compared them with mice with global deficiency in GR (GR(-/-)). Echocardiography shows impaired heart function in both SMGRKO and GR(-/-) mice at embryonic day (E)17.5, associated with generalized oedema. Cardiac ultrastructure is markedly disrupted in both SMGRKO and GR(-/-) mice at E17.5, with short, disorganized myofibrils and cardiomyocytes that fail to align in the compact myocardium. Failure to induce critical genes involved in contractile function, calcium handling and energy metabolism underpins this common phenotype. However, although hearts of GR(-/-) mice are smaller, with 22% reduced ventricular volume at E17.5, SMGRKO hearts are normally sized. Moreover, while levels of mRNA encoding atrial natriuretic peptide are reduced in E17.5 GR(-/-) hearts, they are normal in foetal SMGRKO hearts. These data demonstrate that structural, functional and biochemical maturation of the foetal heart is dependent on glucocorticoid signalling within cardiomyocytes and vascular smooth muscle, though some aspects of heart maturation (size, ANP expression) are independent of GR at these key sites.


Subject(s)
Fetal Heart/growth & development , Glucocorticoids/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction , Animals , Corticosterone/blood , Corticosterone/physiology , Fetal Heart/physiology , Heart/embryology , Heart/physiology , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/embryology , Muscle, Smooth, Vascular/metabolism , Myocardial Contraction , Myocardium/ultrastructure , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myofibrils/ultrastructure
5.
Hum Mol Genet ; 21(18): 3969-83, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22692683

ABSTRACT

Biallelic mutations in the gene encoding DHOdehase [dihydroorotate dehydrogenase (DHODH)], an enzyme required for de novo pyrimidine biosynthesis, have been identified as the cause of Miller (Genée-Weidemann or postaxial acrofacial dysostosis) syndrome (MIM 263750). We report compound heterozygous DHODH mutations in four additional families with typical Miller syndrome. Complementation in auxotrophic yeast demonstrated reduced pyrimidine synthesis and in vitro enzymatic analysis confirmed reduced DHOdehase activity in 11 disease-associated missense mutations, with 7 alleles showing discrepant activity between the assays. These discrepancies are partly explained by the domain structure of DHODH and suggest both assays are useful for interpretation of individual alleles. However, in all affected individuals, the genotype predicts that there should be significant residual DHOdehase activity. Urine samples obtained from two mutation-positive cases showed elevated levels of orotic acid (OA) but not dihydroorotate (DHO), an unexpected finding since these represent the product and the substrate of DHODH enzymatic activity, respectively. Screening of four unrelated cases with overlapping but atypical clinical features showed no mutations in either DHODH or the other de novo pyrimidine biosynthesis genes (CAD, UMPS), with these cases also showing normal levels of urinary OA and DHO. In situ analysis of mouse embryos showed Dhodh, Cad and Umps to be strongly expressed in the pharyngeal arch and limb bud, supporting a site- and stage-specific requirement for de novo pyrimidine synthesis. The developmental sensitivity to reduced pyrimidine synthesis capacity may reflect the requirement for an exceptional mitogenic response to growth factor signalling in the affected tissues.


Subject(s)
Abnormalities, Multiple/enzymology , Limb Deformities, Congenital/enzymology , Mandibulofacial Dysostosis/enzymology , Micrognathism/enzymology , Oxidoreductases Acting on CH-CH Group Donors/deficiency , Abnormalities, Multiple/genetics , Abnormalities, Multiple/urine , Animals , Base Sequence , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/genetics , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Child, Preschool , DNA Mutational Analysis , Dihydroorotate Dehydrogenase , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Gas Chromatography-Mass Spectrometry/standards , Gene Expression Regulation, Developmental , Genetic Association Studies , Genetic Complementation Test , Humans , Infant , Limb Buds/metabolism , Limb Buds/pathology , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/urine , Male , Mandibulofacial Dysostosis/genetics , Mandibulofacial Dysostosis/urine , Mice , Micrognathism/genetics , Micrognathism/urine , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutation, Missense , Orotate Phosphoribosyltransferase/genetics , Orotate Phosphoribosyltransferase/metabolism , Orotic Acid/analogs & derivatives , Orotic Acid/urine , Orotidine-5'-Phosphate Decarboxylase/genetics , Orotidine-5'-Phosphate Decarboxylase/metabolism , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pedigree , Reference Standards , Schizosaccharomyces/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces pombe Proteins/genetics
6.
PLoS Genet ; 7(7): e1002114, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21750680

ABSTRACT

Ophthalmo-acromelic syndrome (OAS), also known as Waardenburg Anophthalmia syndrome, is defined by the combination of eye malformations, most commonly bilateral anophthalmia, with post-axial oligosyndactyly. Homozygosity mapping and subsequent targeted mutation analysis of a locus on 14q24.2 identified homozygous mutations in SMOC1 (SPARC-related modular calcium binding 1) in eight unrelated families. Four of these mutations are nonsense, two frame-shift, and two missense. The missense mutations are both in the second Thyroglobulin Type-1 (Tg1) domain of the protein. The orthologous gene in the mouse, Smoc1, shows site- and stage-specific expression during eye, limb, craniofacial, and somite development. We also report a targeted pre-conditional gene-trap mutation of Smoc1 (Smoc1(tm1a)) that reduces mRNA to ∼10% of wild-type levels. This gene-trap results in highly penetrant hindlimb post-axial oligosyndactyly in homozygous mutant animals (Smoc1(tm1a/tm1a)). Eye malformations, most commonly coloboma, and cleft palate occur in a significant proportion of Smoc1(tm1a/tm1a) embryos and pups. Thus partial loss of Smoc-1 results in a convincing phenocopy of the human disease. SMOC-1 is one of the two mammalian paralogs of Drosophila Pentagone, an inhibitor of decapentaplegic. The orthologous gene in Xenopus laevis, Smoc-1, also functions as a Bone Morphogenic Protein (BMP) antagonist in early embryogenesis. Loss of BMP antagonism during mammalian development provides a plausible explanation for both the limb and eye phenotype in humans and mice.


Subject(s)
Anophthalmos/genetics , Bone Morphogenetic Protein 1/antagonists & inhibitors , Mutation , Osteonectin , Waardenburg Syndrome/genetics , Animals , Bone Morphogenetic Protein 1/genetics , Coloboma/genetics , DNA Mutational Analysis , Extremities/growth & development , Eye/growth & development , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Osteonectin/genetics , Osteonectin/metabolism , Pedigree , Syndactyly/genetics , Xenopus laevis
7.
Hum Mol Genet ; 20(11): 2182-94, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21402589

ABSTRACT

Nephrin (NPHS1) has been described as an important structural protein of kidney podocytes. Mutations in this gene lead to the Finnish-type congenital nephrotic syndrome. More recently, a role of nephrin as a signalling molecule in kidney podocytes has been identified. Here, we show that nephrin not only has a function in kidney podocytes, but is also required for cardiovascular development. Nephrin is expressed in the epicardium and coronary vessels during human and mouse embryonic development. Nephrin knockout embryos showed abnormal epicardial cell morphology and, at later stages of development, a reduced number of coronary vessels due to increased apoptosis, and in addition, cardiac fibrosis. Connexin 43, which is required for coronary vessel formation, was downregulated in nephrin knockout embryos. Expression of the p75NTR neurotrophin receptor, a known mediator of apoptosis, was increased in mutants. Furthermore, co-immunoprecipitation studies demonstrated a direct interaction of nephrin with p75NTR. Primary nephrin-deficient cardiac cells showed a 5-fold higher rate of apoptosis in response to progenitor of nerve growth factor compared with wild-type cells, which could be rescued by RNAi against p75NTR. Taken together, our data demonstrate that nephrin directly interacts with p75NTR and reveal an important role for nephrin in murine cardiac development by permitting survival of cardiovascular progenitor cells.


Subject(s)
Coronary Vessels/embryology , Membrane Proteins/metabolism , Pericardium/embryology , Podocytes/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Apoptosis , Connexin 43/genetics , Connexin 43/metabolism , Coronary Vessels/metabolism , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Humans , Immunoprecipitation , Kidney/cytology , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Organogenesis/genetics , Pericardium/metabolism , Receptors, Nerve Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Two-Hybrid System Techniques
8.
Dev Dyn ; 241(2): 340-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22113860

ABSTRACT

BACKGROUND: DNA variation in Interferon Regulatory Factor 6 (IRF6) contributes risk for orofacial clefting, including a common DNA variant rs642961. This DNA variant is located in a multi-species conserved sequence that is 9.7 kb upstream from the IRF6 transcriptional start site (MCS9.7). The MCS9.7 element was shown to possess enhancer activity that mimicked the expression of endogenous Irf6 at embryonic day 11.5 in transient transgenic embryos, and also contains a p63 binding site that transactivates IRF6 expression. To analyze whether the MCS9.7 enhancer is sufficient to drive IRF6 expression, we generated stable transgenic murine lines that carry a MCS9.7-lacZ transgene. We hypothesized that MCS9.7 was sufficient to recapitulate the endogenous expression of Irf6 at other time-points during embryonic development. RESULTS: We observed that MCS9.7 activity recapitulated endogenous Irf6 expression in most tissues, but not in the medial edge epithelium (MEE) at E14.5, when Irf6 expression was high during secondary palatal fusion. Also, while MCS9.7 activity and Irf6 expression were associated with p63 expression, we observed MCS9.7 activity and Irf6 expression in periderm, although p63 was absent. CONCLUSION: These data suggest that MCS9.7 enhancer activity is not sufficient to recapitulate IRF6 expression, and that p63 expression is not always necessary nor sufficient for transactivation of IRF6.


Subject(s)
Enhancer Elements, Genetic , Epidermis/embryology , Gene Expression Regulation, Developmental , Interferon Regulatory Factors/genetics , Palate/embryology , Phosphoproteins/genetics , Trans-Activators/genetics , Transcriptional Activation , Animals , Cleft Lip/genetics , Cleft Palate/genetics , Epithelium/embryology , Mice , Mice, Transgenic , Palate/metabolism , Transcription Initiation Site , beta-Galactosidase/genetics
9.
Hum Mutat ; 32(12): 1492-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21948517

ABSTRACT

Disruption of the long-range cis-regulation of developmental gene expression is increasingly recognized as a cause of human disease. Here, we report a novel type of long-range cis-regulatory mutation, in which ectopic expression of a gene is driven by an enhancer that is not its own. We have termed this gain of regulatory information as "enhancer adoption." We mapped the breakpoints of a de novo 7q inversion in a child with features of a holoprosencephaly spectrum (HPES) disorder and severe upper limb syndactyly with lower limb synpolydactyly. The HPES plausibly results from the 7q36.3 breakpoint dislocating the sonic hedgehog (SHH) gene from enhancers that are known to drive expression in the early forebrain. However, the limb phenotype cannot be explained by loss of known SHH enhancers. The SHH transcription unit is relocated to 7q22.1, ∼190 kb 3' of a highly conserved noncoding element (HCNE2) within an intron of EMID2. We show that HCNE2 functions as a limb bud enhancer in mouse embryos and drives ectopic expression of Shh in vivo recapitulating the limb phenotype in the child. This developmental genetic mechanism may explain a proportion of the novel or unexplained phenotypes associated with balanced chromosome rearrangements.


Subject(s)
Chromosome Inversion/genetics , Enhancer Elements, Genetic/genetics , Hedgehog Proteins/genetics , Holoprosencephaly/genetics , Syndactyly/genetics , Animals , Child, Preschool , Chromosomes, Human, Pair 7/genetics , Extremities/embryology , Female , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Humans , Limb Buds/embryology , Mice , Mice, Transgenic , Mutation
10.
Mol Cell Neurosci ; 37(4): 857-68, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18329286

ABSTRACT

We used Fgf10-lacZ reporter mice to investigate the distribution and fate of Fgf10-expressing cells in the developing and adult mouse brain. We find that the domain of Fgf10 expression expands post-natally and new niches emerge in the adult brain. Fgf10 is expressed in the adult cerebellum, thalamic, mid- and hindbrain nuclei and hippocampal CA fields, as previously reported in the rat brain. In addition though, we have discovered expression in: the hippocampal dentate gyrus; a discrete trail linking the ventral telencephalon with the olfactory bulbs; ventral ependyma of the third ventricle from where cells appear to disperse into the hypothalamus; and in the pituitary gland. Most Fgf10-expressing cells or their immediate descendants appear immature but a subset differentiates into neurons and glial cells. The manner in which Fgf10 is expressed in these active and quiescent neurogenic niches implicates it in control of neurogenesis and/or conservation of neurogenic potential.


Subject(s)
Brain/growth & development , Cell Differentiation/physiology , Fibroblast Growth Factor 10/genetics , Gene Expression Regulation, Developmental/physiology , Neurons/cytology , Neurons/physiology , Age Factors , Animals , Brain/cytology , Fibroblast Growth Factor 10/biosynthesis , Fibroblast Growth Factor 10/physiology , Mice , Mice, Transgenic , Neurons/chemistry
11.
Cell Rep ; 20(8): 1745-1754, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28834739

ABSTRACT

Coronary arteries are essential to support the heart with oxygen, and coronary heart disease is one of the leading causes of death worldwide. The coronary arteries form at highly stereotyped locations and are derived from the primitive vascular plexus of the heart. How coronary arteries are remodeled and the signaling molecules that govern this process are poorly understood. Here, we have identified the Wnt-signaling modulator Rspo3 as a crucial regulator of coronary artery formation in the developing heart. Rspo3 is specifically expressed around the coronary stems at critical time points in their development. Temporal ablation of Rspo3 at E11.5 leads to decreased ß-catenin signaling and a reduction in arterial-specific proliferation. As a result, the coronary stems are defective and the arterial tree does not form properly. These results identify a mechanism through which localized expression of RSPO3 induces proliferation of the coronary arteries at their stems and permits their formation.


Subject(s)
Coronary Vessels/growth & development , Coronary Vessels/metabolism , Thrombospondins/biosynthesis , Animals , Cell Proliferation/physiology , Female , Mice , Neovascularization, Physiologic/physiology , Pregnancy , Wnt Signaling Pathway
12.
Dis Model Mech ; 7(6): 711-22, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24764192

ABSTRACT

Mutations in RAB18 have been shown to cause the heterogeneous autosomal recessive disorder Warburg Micro syndrome (WARBM). Individuals with WARBM present with a range of clinical symptoms, including ocular and neurological abnormalities. However, the underlying cellular and molecular pathogenesis of the disorder remains unclear, largely owing to the lack of any robust animal models that phenocopy both the ocular and neurological features of the disease. We report here the generation and characterisation of a novel Rab18-mutant mouse model of WARBM. Rab18-mutant mice are viable and fertile. They present with congenital nuclear cataracts and atonic pupils, recapitulating the characteristic ocular features that are associated with WARBM. Additionally, Rab18-mutant cells exhibit an increase in lipid droplet size following treatment with oleic acid. Lipid droplet abnormalities are a characteristic feature of cells taken from WARBM individuals, as well as cells taken from individuals with other neurodegenerative conditions. Neurological dysfunction is also apparent in Rab18-mutant mice, including progressive weakness of the hind limbs. We show that the neurological defects are, most likely, not caused by gross perturbations in synaptic vesicle recycling in the central or peripheral nervous system. Rather, loss of Rab18 is associated with widespread disruption of the neuronal cytoskeleton, including abnormal accumulations of neurofilament and microtubule proteins in synaptic terminals, and gross disorganisation of the cytoskeleton in peripheral nerves. Global proteomic profiling of peripheral nerves in Rab18-mutant mice reveals significant alterations in several core molecular pathways that regulate cytoskeletal dynamics in neurons. The apparent similarities between the WARBM phenotype and the phenotype that we describe here indicate that the Rab18-mutant mouse provides an important platform for investigation of the disease pathogenesis and therapeutic interventions.


Subject(s)
Abnormalities, Multiple/physiopathology , Cataract/congenital , Cornea/abnormalities , Cytoskeleton/physiology , Disease Models, Animal , Eye/growth & development , Hypogonadism/physiopathology , Intellectual Disability/physiopathology , Microcephaly/physiopathology , Neurons/physiology , Optic Atrophy/physiopathology , rab GTP-Binding Proteins/physiology , Animals , Cataract/physiopathology , Cornea/physiopathology , Mice , Mice, Knockout , rab GTP-Binding Proteins/genetics
13.
Science ; 342(6157): 1241006, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24159046

ABSTRACT

The shape of the human face and skull is largely genetically determined. However, the genomic basis of craniofacial morphology is incompletely understood and hypothesized to involve protein-coding genes, as well as gene regulatory sequences. We used a combination of epigenomic profiling, in vivo characterization of candidate enhancer sequences in transgenic mice, and targeted deletion experiments to examine the role of distant-acting enhancers in craniofacial development. We identified complex regulatory landscapes consisting of enhancers that drive spatially complex developmental expression patterns. Analysis of mouse lines in which individual craniofacial enhancers had been deleted revealed significant alterations of craniofacial shape, demonstrating the functional importance of enhancers in defining face and skull morphology. These results demonstrate that enhancers are involved in craniofacial development and suggest that enhancer sequence variation contributes to the diversity of human facial morphology.


Subject(s)
Enhancer Elements, Genetic/physiology , Face/anatomy & histology , Gene Expression Regulation, Developmental , Maxillofacial Development/genetics , Skull/growth & development , Animals , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Face/abnormalities , Gene Expression Profiling , Gene Targeting , Mice , Mice, Transgenic , Sequence Deletion , Skull/abnormalities , Skull/anatomy & histology
14.
Science ; 321(5894): 1346-50, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18772437

ABSTRACT

Changes in gene regulation are thought to have contributed to the evolution of human development. However, in vivo evidence for uniquely human developmental regulatory function has remained elusive. In transgenic mice, a conserved noncoding sequence (HACNS1) that evolved extremely rapidly in humans acted as an enhancer of gene expression that has gained a strong limb expression domain relative to the orthologous elements from chimpanzee and rhesus macaque. This gain of function was consistent across two developmental stages in the mouse and included the presumptive anterior wrist and proximal thumb. In vivo analyses with synthetic enhancers, in which human-specific substitutions were introduced into the chimpanzee enhancer sequence or reverted in the human enhancer to the ancestral state, indicated that 13 substitutions clustered in an 81-base pair module otherwise highly constrained among terrestrial vertebrates were sufficient to confer the human-specific limb expression domain.


Subject(s)
Body Patterning/genetics , Enhancer Elements, Genetic , Extremities/embryology , Gene Expression Regulation, Developmental , Animals , Base Sequence , Binding Sites , Conserved Sequence , Embryonic Development , Evolution, Molecular , Gene Expression Profiling , Humans , Limb Buds/embryology , Limb Buds/metabolism , Macaca mulatta/genetics , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , PAX9 Transcription Factor/metabolism , Pan troglodytes/genetics , Selection, Genetic , Transcription Factors/metabolism
15.
Nat Methods ; 4(1): 31-3, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17143281

ABSTRACT

A convenient technology to quantify three-dimensional (3D) morphological features would have widespread applications in biomedical research. Based on combined improvements in sample preparation, tomographic imaging and computational processing, we present a procedure for high-resolution 3D quantification of structures within intact adult mouse organs. Using the nonobese diabetic (NOD) mouse model, we demonstrate a correlation between total islet beta-cell volume and the onset of type-1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/diagnosis , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Islets of Langerhans/pathology , Pancreas/pathology , Tomography/methods , Animals , Diabetes Mellitus, Type 1/physiopathology , Disease Models, Animal , Disease Progression , Mice , Sensitivity and Specificity
16.
PLoS One ; 2(9): e834, 2007 Sep 05.
Article in English | MEDLINE | ID: mdl-17786206

ABSTRACT

BACKGROUND: The major hindrance to imaging the intact adult Drosophila is that the dark exoskeleton makes it impossible to image through the cuticle. We have overcome this obstacle and describe a method whereby the internal organs of adult Drosophila can be imaged in 3D by bleaching and clearing the adult and then imaging using a technique called optical projection tomography (OPT). The data is displayed as 2D optical sections and also in 3D to provide detail on the shape and structure of the adult anatomy. METHODOLOGY: We have used OPT to visualize in 2D and 3D the detailed internal anatomy of the intact adult Drosophila. In addition this clearing method used for OPT was tested for imaging with confocal microscopy. Using OPT we have visualized the size and shape of neurodegenerative vacuoles from within the head capsule of flies that suffer from age-related neurodegeneration due to a lack of ADAR mediated RNA-editing. In addition we have visualized tau-lacZ expression in 2D and 3D. This shows that the wholemount adult can be stained without any manipulation and that this stain penetrates well as we have mapped the localization pattern with respect to the internal anatomy. CONCLUSION: We show for the first time that the intact adult Drosophila can be imaged in 3D using OPT, also we show that this method of clearing is also suitable for confocal microscopy to image the brain from within the intact head. The major advantage of this is that organs can be represented in 3D in their natural surroundings. Furthermore optical sections are generated in each of the three planes and are not prone to the technical limitations that are associated with manual sectioning. OPT can be used to dissect mutant phenotypes and to globally map gene expression in both 2D and 3D.


Subject(s)
Drosophila melanogaster/anatomy & histology , Imaging, Three-Dimensional , Animals , Microscopy, Confocal
17.
Plant Cell ; 18(9): 2145-56, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16905654

ABSTRACT

A deeper understanding of the mechanisms that underlie plant growth and development requires quantitative data on three-dimensional (3D) morphology and gene activity at a variety of stages and scales. To address this, we have explored the use of optical projection tomography (OPT) as a method for capturing 3D data from plant specimens. We show that OPT can be conveniently applied to a wide variety of plant material at a range of scales, including seedlings, leaves, flowers, roots, seeds, embryos, and meristems. At the highest resolution, large individual cells can be seen in the context of the surrounding plant structure. For naturally semitransparent structures, such as roots, live 3D imaging using OPT is also possible. 3D domains of gene expression can be visualized using either marker genes, such as beta-glucuronidase, or more directly by whole-mount in situ hybridization. We also describe tools and software that allow the 3D data to be readily quantified and visualized interactively in different ways.


Subject(s)
Plant Development , Plant Proteins/genetics , Plants/embryology , Tomography, Optical/methods , Flowers/anatomy & histology , Flowers/metabolism , Flowers/physiology , Gene Expression , Genes, Reporter , Glucuronidase/analysis , Imaging, Three-Dimensional/methods , Meristem/anatomy & histology , Meristem/metabolism , Meristem/physiology , Plant Leaves/anatomy & histology , Plant Leaves/metabolism , Plant Leaves/physiology , Plant Proteins/metabolism , Plant Roots/anatomy & histology , Plant Roots/metabolism , Plant Roots/physiology , Plants/anatomy & histology , Seedlings/anatomy & histology , Seedlings/metabolism , Seedlings/physiology , Seeds/anatomy & histology , Seeds/metabolism , Seeds/physiology , Software
18.
Dev Biol ; 283(1): 113-27, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15893304

ABSTRACT

Null mutation of the Foxg1 gene causes hypoplasia of the mouse telencephalon and loss of ventral telencephalic structures. We show that a crucial early requirement for Foxg1 is in the induction of ventral cell fate in the telencephalon. To study later proliferative defects, we have adapted an iododeoxyuridine and bromodeoxyuridine double labeling protocol for use in the developing embryo, which allows estimation of cell cycle kinetics in a single specimen. This technique is used to demonstrate that the cell cycle is prematurely lengthened in the Foxg1-null telencephalon. These defects are first apparent at embryonic day 10.5 (E10.5) and are most severe in the rostral telencephalon. We show that apoptosis is also reduced in the same rostral domain. These defects correspond temporally and spatially with a dramatic reduction in expression of the potent signaling molecule Fgf8. We also show that in the absence of Foxg1 an excess of neurons is produced from E11.5, depleting the progenitor pool and limiting the growth of the Foxg1(-/-) telencephalon. The increase in neurogenic division coincides with an increase in BMP signaling, as detected by immunohistochemistry for phosphorylated smad-1, -5, and -8. This study reinforces Foxg1's position as a major regulator of telencephalic neurogenesis and supports the idea that Foxg1 controls precursor proliferation via regulation of Fgf signaling and differentiation via regulation of Bmp signaling.


Subject(s)
DNA-Binding Proteins/genetics , Nerve Tissue Proteins/genetics , Telencephalon/embryology , Animals , Apoptosis , Cell Cycle , Cell Division , DNA-Binding Proteins/deficiency , Embryonic Development , Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Kinetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Nerve Tissue Proteins/deficiency , S Phase , Telencephalon/cytology , Telencephalon/physiology , Transcription Factors/genetics
19.
Chromosome Res ; 10(7): 579-95, 2002.
Article in English | MEDLINE | ID: mdl-12498347

ABSTRACT

We have used a novel approach to investigate the histone H4 acetylation status at X-inactivated genes compared with their active counterparts. Immunoprecipitation with a sheep antibody that preferentially binds multiply-acetylated H4 isoforms was used to select hyperacetylated chromatin from a human female lymphoblastoid cell line exhibiting non-random X-inactivation as a result of an X/autosome translocation. The distribution of active and inactive gene sequences between the immunoprecipitated and bulk chromatin was compared at four X-linked loci containing intragenic polymorphic microsatellite repeats to allow identification of individual alleles by polymerase chain reaction. We find that DNA sequences corresponding to transcriptionally silent alleles are consistently under-represented in the hyperacetylated fraction. As the microsatellite repeat sequences used to identify alleles range in distance from 6.5 kb to 25 kb downstream of promoters, we conclude that differential H4 acetylation of active and silent chromatin is not confined to regions involved in the initiation of transcription, contrary to previous reports.


Subject(s)
Alleles , Dosage Compensation, Genetic , Histones/metabolism , Promoter Regions, Genetic , Acetylation , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromosome Painting , DNA/genetics , DNA/metabolism , Female , Genetic Markers , Humans , Microsatellite Repeats , Nucleosomes/genetics , Nucleosomes/metabolism , Polymorphism, Genetic , Protein Isoforms/metabolism , Translocation, Genetic
20.
Chromosoma ; 111(1): 22-36, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12068920

ABSTRACT

We show that methylated lysine 9 of histone H3 (Me9H3) is a marker of heterochromatin in divergent animal species. It localises to both constitutive and facultative heterochromatin and replicates late in S-phase of the cell cycle. Significantly, Me9H3 is enriched in the inactive mammalian X chromosome (Xi) in female cells, as well as in the XY body during meiosis in the male, and forms a G-band pattern along the arms of the autosomes. Me9H3 is a constituent of imprinted chromosomes that are repressed. The paternal and maternal pronuclei in one-cell mouse embryos show a striking non-equivalence in Me9H3: the paternal pronucleus contains no immunocytologically detectable Me9H3. The levels of Me9H3 on the parental chromosomes only become equivalent after the two-cell stage. Finally, we provide evidence that Me9H3 is neither necessary nor sufficient for localisation of heterochromatin protein 1 (HP1) to chromosomal DNA.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Heterochromatin/metabolism , Histones/metabolism , Lysine/metabolism , Animals , Bisbenzimidazole , Chromobox Protein Homolog 5 , DNA/metabolism , Drosophila/genetics , Drosophila/metabolism , Female , Fluorescent Antibody Technique , Gene Silencing/physiology , Genomic Imprinting , Male , Methylation , Mice , Microscopy, Fluorescence , Oocytes/metabolism , Spermatozoa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL