Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Exp Anim ; 73(3): 310-318, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38447983

ABSTRACT

Allele-specific monoallelic gene expression is a unique phenomenon and a great resource for analyzing gene regulation. To study this phenomenon, we established new embryonic stem (ES) cell lines derived from F1 hybrid blastocysts from crosses between four mouse subspecies (Mus musculus domesticus, C57BL/6; M. musculus molossinus, MSM/Ms; M. musculus musculus, PWK; M. musculus castaneus, HMI/Ms) and analyzed the expression levels of undifferentiated pluripotent stem cell markers and karyotypes of each line. To demonstrate the utility of our cell lines, we analyzed the allele-specific expression pattern of the Inpp5d gene as an example. The allelic expression depended on the parental alleles; this dependence could be a consequence of differences in compatibility between cis- and trans-elements of the Inpp5d gene from different subspecies. The use of parental mice from four subspecies greatly enhanced genetic polymorphism. The F1 hybrid ES cells retained this polymorphism not only in the Inpp5d gene, but also at a genome-wide level. As we demonstrated for the Inpp5d gene, the established cell lines can contribute to the analysis of allelic expression imbalance based on the incompatibility between cis- and trans-elements and of phenotypes related to this incompatibility.


Subject(s)
Allelic Imbalance , Animals , Mice , Allelic Imbalance/genetics , Mice, Inbred C57BL , Alleles , Gene Expression/genetics , Cell Line , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Polymorphism, Genetic , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Hybrid Cells , Embryonic Stem Cells , Female , Species Specificity , Male
2.
Dev Biol ; 500: 22-30, 2023 08.
Article in English | MEDLINE | ID: mdl-37247832

ABSTRACT

Xenopus young tadpoles regenerate a limb with the anteroposterior (AP) pattern, but metamorphosed froglets regenerate a hypomorphic limb after amputation. The key gene for AP patterning, shh, is expressed in a regenerating limb of the tadpole but not in that of the froglet. Genomic DNA in the shh limb-specific enhancer, MFCS1 (ZRS), is hypermethylated in froglets but hypomethylated in tadpoles: shh expression may be controlled by epigenetic regulation of MFCS1. Is MFCS1 specifically activated for regenerating the AP-patterned limb? We generated transgenic Xenopus laevis lines that visualize the MFCS1 enhancer activity with a GFP reporter. The transgenic tadpoles showed GFP expression in hoxd13-and shh-expressing domains of developing and regenerating limbs, whereas the froglets showed no GFP expression in the regenerating limbs despite having hoxd13 expression. Genome sequence analysis and co-transfection assays using cultured cells revealed that Hoxd13 can activate Xenopus MFCS1. These results suggest that MFCS1 activation correlates with regeneration of AP-patterned limbs and that re-activation of epigenetically inactivated MFCS1 would be crucial to confer the ability to non-regenerative animals for regenerating a properly patterned limb.


Subject(s)
Epigenesis, Genetic , Extremities , Animals , Xenopus laevis/genetics , Animals, Genetically Modified , Extremities/physiology , Transcription Factors/genetics
3.
Carcinogenesis ; 44(4): 341-349, 2023 06 24.
Article in English | MEDLINE | ID: mdl-36753047

ABSTRACT

Chronic inflammation is widely recognized as a major risk factor for cancer formation, but the underlying mechanisms are poorly understood. Recently, it was shown that Gasdermin D (GSDMD) protein drives pyroptotic cell death in macrophages on cleavage by inflammatory caspases. Even though the Gsdmd gene is specifically expressed in the intestinal epithelium, the role of Gsdmd in the intestinal tissues remains poorly characterized. In this study, we examined the biological role of Gsdmd in colorectal cancer (CRC) development, employing an azoxymethane/dextran sulfate sodium carcinogenesis model. Results show that GSDMD deficiency enhances CRC development, probably due to decreased apoptosis caused by downregulation of interferon-gamma (IFNγ)-signal transducer and activator 1 (STAT1) signaling. Furthermore, we show that GSDMD protein is diminished in human colorectal cancer, indicating involvement of GSDMD in repression of CRC development in humans. Our findings provide a new insight into functions of Gsdmd/GSDMD in colonic inflammation and human CRC development.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Humans , Gasdermins , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/genetics , Apoptosis , Inflammation , Colonic Neoplasms/genetics
4.
Dev Growth Differ ; 62(5): 334-342, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32343848

ABSTRACT

The organs of vertebrate species display a wide variety of morphology. A remaining challenge in evolutionary developmental biology is to elucidate how vertebrate lineages acquire distinct morphological features. Developmental programs are driven by spatiotemporal regulation of gene expression controlled by hundreds of thousands of cis-regulatory elements. Changes in the regulatory elements caused by the introduction of genetic variants can confer regulatory innovation that may underlie morphological novelties. Recent advances in sequencing technology have revealed a number of potential regulatory variants that can alter gene expression patterns. However, a limited number of studies demonstrate causal dependence between genetic and morphological changes. Regulation of Shh expression is a good model to understand how multiple regulatory elements organize tissue-specific gene expression patterns. This model also provides insights into how evolution of molecular traits, such as gene regulatory networks, lead to phenotypic novelty.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Hedgehog Proteins/genetics , Animals , Enhancer Elements, Genetic/genetics , Hedgehog Proteins/metabolism , Humans , Phenotype
5.
Development ; 146(21)2019 11 12.
Article in English | MEDLINE | ID: mdl-31719045

ABSTRACT

The tongue is a highly specialised muscular organ with a complex anatomy required for normal function. We have utilised multiple genetic approaches to investigate local temporospatial requirements for sonic hedgehog (SHH) signalling during tongue development. Mice lacking a Shh cis-enhancer, MFCS4 (ShhMFCS4/-), with reduced SHH in dorsal tongue epithelium have perturbed lingual septum tendon formation and disrupted intrinsic muscle patterning, with these defects reproduced following global Shh deletion from E10.5 in pCag-CreERTM; Shhflox/flox embryos. SHH responsiveness was diminished in local cranial neural crest cell (CNCC) populations in both mutants, with SHH targeting these cells through the primary cilium. CNCC-specific deletion of orofaciodigital syndrome 1 (Ofd1), which encodes a ciliary protein, in Wnt1-Cre; Ofdfl/Y mice led to a complete loss of normal myotube arrangement and hypoglossia. In contrast, mesoderm-specific deletion of Ofd1 in Mesp1-Cre; Ofdfl/Y embryos resulted in normal intrinsic muscle arrangement. Collectively, these findings suggest key temporospatial requirements for local SHH signalling in tongue development (specifically, lingual tendon differentiation and intrinsic muscle patterning through signalling to CNCCs) and provide further mechanistic insight into the tongue anomalies seen in patients with disrupted hedgehog signalling.


Subject(s)
Body Patterning , Hedgehog Proteins/metabolism , Neural Crest/cytology , Signal Transduction , Tongue/embryology , Alleles , Animals , Cell Proliferation , Enhancer Elements, Genetic , Female , Gene Deletion , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Heterozygote , Ligands , Mesoderm/metabolism , Mice , Morphogenesis/genetics , Phenotype , Proteins/metabolism , Tendons/metabolism , Time Factors , Transforming Growth Factor beta/metabolism , Wnt1 Protein/metabolism
6.
Proc Natl Acad Sci U S A ; 116(47): 23636-23642, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31685615

ABSTRACT

Sonic hedgehog (SHH) signaling plays a pivotal role in 2 different phases during brain development. Early SHH signaling derived from the prechordal plate (PrCP) triggers secondary Shh induction in the forebrain, which overlies the PrCP, and the induced SHH signaling, in turn, directs late neuronal differentiation of the forebrain. Consequently, Shh regulation in the PrCP is crucial for initiation of forebrain development. However, no enhancer that regulates prechordal Shh expression has yet been found. Here, we identified a prechordal enhancer, named SBE7, in the vicinity of a cluster of known forebrain enhancers for Shh This enhancer also directs Shh expression in the ventral midline of the forebrain, which receives the prechordal SHH signal. Thus, the identified enhancer acts not only for the initiation of Shh regulation in the PrCP but also for subsequent Shh induction in the forebrain. Indeed, removal of the enhancer from the mouse genome markedly down-regulated the expression of Shh in the rostral domains of the axial mesoderm and in the ventral midline of the forebrain and hypothalamus in the mouse embryo, and caused a craniofacial abnormality similar to human holoprosencephaly (HPE). These findings demonstrate that SHH signaling mediated by the newly identified enhancer is essential for development and growth of the ventral midline of the forebrain and hypothalamus. Understanding of the Shh regulation governed by this prechordal and brain enhancer provides an insight into the mechanism underlying craniofacial morphogenesis and the etiology of HPE.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Hedgehog Proteins/physiology , Nerve Tissue Proteins/physiology , Prosencephalon/embryology , Animals , CRISPR-Cas Systems , Eye Proteins/physiology , Gene Knockout Techniques , Genes, Reporter , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/genetics , Holoprosencephaly/genetics , Homeodomain Proteins/physiology , Hypothalamus/abnormalities , Hypothalamus/embryology , Hypothalamus/metabolism , Lac Operon , Mesencephalon/embryology , Mesencephalon/metabolism , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Prosencephalon/abnormalities , Prosencephalon/metabolism , Signal Transduction , Transgenes , Homeobox Protein SIX3
7.
Sci Rep ; 8(1): 9585, 2018 06 25.
Article in English | MEDLINE | ID: mdl-29941919

ABSTRACT

Accumulation of ubiquitinated protein aggregates is a common pathology associated with a number of neurodegenerative diseases and selective autophagy plays a critical role in their elimination. Although aging-related decreases in protein degradation properties may enhance protein aggregation, it remains unclear whether proteasome dysfunction is indispensable for ubiquitinated-protein aggregation in neurodegenerative diseases. Here, we show that N-oleoyl-dopamine and N-arachidonyl-dopamine, which are endogenous brain substances and belong to the N-acyldopamine (AcylDA) family, generate cellular inclusions through aggresome formation without proteasome inhibition. Although AcylDA itself does not inhibit proteasome activity in vitro, it activates the rearrangement of vimentin distribution to form a vimentin cage surrounding aggresomes and sequesters ubiquitinated proteins in aggresomes. The gene transcription of p62/SQSTM1 was significantly increased by AcylDAs, whereas the transcription of other ubiquitin-dependent autophagy receptors was unaffected. Genetic depletion of p62 resulted in the loss of ubiquitinated-protein sequestration in aggresomes, indicating that p62 is a critical component of aggresomes. Furthermore, AcylDAs accelerate the aggregation of mutant huntingtin exon 1 proteins. These results suggest that aggresome formation does not require proteasome dysfunction and AcylDA-induced aggresome formation may participate in forming cytoplasmic protein inclusions.


Subject(s)
Arachidonic Acids/metabolism , Dopamine/analogs & derivatives , Gene Expression Regulation/drug effects , Protein Aggregates/drug effects , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Arachidonic Acids/pharmacology , Autophagy/drug effects , Cell Line , Dopamine/metabolism , Dopamine/pharmacology , Drug Evaluation, Preclinical , Humans , Huntingtin Protein/chemistry , Huntingtin Protein/genetics , Leupeptins/pharmacology , Mutation , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Transcription, Genetic/drug effects
8.
Proc Natl Acad Sci U S A ; 115(5): 1021-1026, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29255029

ABSTRACT

Acquisition of new cis-regulatory elements (CREs) can cause alteration of developmental gene regulation and may introduce morphological novelty in evolution. Although structural variation in the genome generated by chromosomal rearrangement is one possible source of new CREs, only a few examples are known, except for cases of retrotransposition. In this study, we show the acquisition of novel regulatory sequences as a result of large genomic insertion in the spontaneous mouse mutation Hammer toe (Hm). Hm mice exhibit syndactyly with webbing, due to suppression of interdigital cell death in limb development. We reveal that, in the Hm genome, a 150-kb noncoding DNA fragment from chromosome 14 is inserted into the region upstream of the Sonic hedgehog (Shh) promoter in chromosome 5. Phenotyping of mouse embryos with a series of CRISPR/Cas9-aided partial deletion of the 150-kb insert clearly indicated that two different regions are necessary for the syndactyly phenotype of Hm We found that each of the two regions contains at least one enhancer for interdigital regulation. These results show that a set of enhancers brought by the large genomic insertion elicits the interdigital Shh expression and the Hm phenotype. Transcriptome analysis indicates that ectopic expression of Shh up-regulates Chordin (Chrd) that antagonizes bone morphogenetic protein signaling in the interdigital region. Indeed, Chrd-overexpressing transgenic mice recapitulated syndactyly with webbing. Thus, the Hm mutation provides an insight into enhancer acquisition as a source of creation of novel gene regulation.


Subject(s)
Enhancer Elements, Genetic , Hedgehog Proteins/genetics , Syndactyly/genetics , Animals , Gene Expression Regulation, Developmental , Genetic Linkage , Glycoproteins/genetics , Hedgehog Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Mutant Strains , Mice, Transgenic , Mutagenesis, Insertional , Mutation , Phenotype , Syndactyly/embryology , Syndactyly/metabolism
9.
Sci Rep ; 7(1): 13004, 2017 10 11.
Article in English | MEDLINE | ID: mdl-29021530

ABSTRACT

Interaction between the epithelium and mesenchyme coordinates patterning and differentiation of oral cavity structures including teeth, palatal rugae and tongue papillae. SHH is one of the key signaling molecules for this interaction. Epithelial expression of Shh in the tooth buds and tongue papillae is regulated by at least two enhancers, MRCS1 and MFCS4. However, it is unclear how the two enhancers cooperate to regulate Shh. Here, we found that simultaneous deletion of MRCS1 and MFCS4 results in the formation of a supernumerary tooth in front of the first molar. Since deletion of either single enhancer barely affects tooth development, MRCS1 and MFCS4 evidently act in a redundant fashion. Binding motifs for WNT signaling mediators are shared by MRCS1 and MFCS4, and play a central role in regulating Shh expression, indicating that the two redundant enhancers additively exert their Shh regulation by responding to WNT signal input.


Subject(s)
Enhancer Elements, Genetic/genetics , Epithelium/embryology , Epithelium/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Tooth/embryology , Tooth/metabolism , Animals , Base Sequence , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mice, Knockout , Nucleotide Motifs/genetics , Sequence Deletion , Tooth, Supernumerary/genetics , Xenopus/genetics
10.
G3 (Bethesda) ; 7(9): 2991-2998, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28710291

ABSTRACT

An enhancer named MFCS1 regulates Sonic hedgehog (Shh) expression in the posterior mesenchyme of limb buds. Several mutations in MFCS1 induce ectopic Shh expression in the anterior limb bud, and these result in preaxial polydactyly (PPD). However, the molecular basis of ectopic Shh expression remains elusive, although some mutations are known to disrupt the negative regulation of Shh expression in the anterior limb bud. Here, we analyzed the molecular mechanism of ectopic Shh expression in PPD including in a mouse mutation-hemimelic extra toes (Hx)-and in other MFCS1 mutations in different species. First, we generated transgenic mouse lines with a LacZ reporter cassette flanked with tandem repeats of 40 bp MFCS1 fragments harboring a mutation. The transgenic mouse line with the Hx-type fragment showed reporter expression exclusively in the anterior, but not in the posterior margins of limb buds. In contrast, no specific LacZ expression was observed in lines carrying the MFCS1 fragment with other mutations. Yeast one-hybrid assays revealed that the msh-like homeodomain protein, MSX1, bound specifically to the Hx sequence of MFCS1. Thus, PPD caused by mutations in MFCS1 has two major types of molecular etiology: loss of a cis-motif for negative regulation of Shh, and acquisition of a new cis-motif binding to a preexisting transcription factor, as represented by the Hx mutation.


Subject(s)
Body Patterning/genetics , Enhancer Elements, Genetic , Extremities/embryology , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Mutation , Animals , Base Sequence , Binding Sites , Ectopic Gene Expression , Gene Expression , Genes, Reporter , Mice , Mice, Transgenic , Organ Specificity/genetics , Phenotype , Point Mutation , Protein Binding
11.
Nat Commun ; 8: 14300, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28155855

ABSTRACT

Shh signalling plays a crucial role for endoderm development. A Shh endoderm enhancer, MACS1, is well conserved across terrestrial animals with lungs. Here, we first show that eliminating mouse MACS1 causes severe defects in laryngeal development, indicating that MACS1-directed Shh signalling is indispensable for respiratory organogenesis. Extensive phylogenetic analyses revealed that MACS1 emerged prior to the divergence of cartilaginous and bony fishes, and even euteleost fishes have a MACS1 orthologue. Meanwhile, ray-finned fishes evolved a novel conserved non-coding sequence in the neighbouring region. Transgenic assays showed that MACS1 drives reporter expression ventrally in laryngeal epithelium. This activity has been lost in the euteleost lineage, and instead, the conserved non-coding sequence of euteleosts acquired an enhancer activity to elicit dorsal epithelial expression in the posterior pharynx and oesophagus. These results implicate that evolution of these two enhancers is relevant to the morphological transition from ventral lungs to dorsal gas bladder.


Subject(s)
Air Sacs/embryology , Enhancer Elements, Genetic , Evolution, Molecular , Hedgehog Proteins/genetics , Lung/embryology , Animals , Animals, Genetically Modified , Binding Sites , Coenzyme A Ligases/genetics , Fishes/embryology , Fishes/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , Introns , Larynx/embryology , Larynx/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Oryzias , Phylogeny , Regulatory Sequences, Nucleic Acid , Signal Transduction
12.
Curr Top Dev Biol ; 110: 129-52, 2014.
Article in English | MEDLINE | ID: mdl-25248475

ABSTRACT

Heart- and neural crest derivatives-expressed (Hand) proteins belong to the Twist family of the basic helix-loop-helix (bHLH) transcription factors, and play crucial roles in the development of several organs. They form heterodimers with Twist1 via their HLH domain. Disruption of the expression balance between Hand2 and Twist1 causes limb malformation, indicating that the expression level of Hand2 relative to Twist1 is essential for limb development. Mutations of the TWIST1 and TWIST2 genes are involved in human diseases. Although, the functions of the Hand proteins are indispensable for limb, heart, and craniofacial development, mutations of the Hand genes that are causative of human diseases remain elusive. Recently, comparative analyses of a human chromosomal disorder, partial trisomy distal 4q, and its mouse model, which is a spontaneously occurring mutant, clearly demonstrated that over dosage of Hand2 results in developmental defects of limbs, craniofacial, and lumbar vertebrae, and that trisomy of the Hand2 gene directly causes a human congenital disorder. In this review, we focus on gene dosage effect of Hand2 in limb, heart, and craniofacial development, and discuss its implication in human diseases.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Congenital Abnormalities/genetics , Gene Dosage , Gene Expression Regulation, Developmental , Amino Acid Sequence , Animals , Chromosomes, Human, Pair 4 , Embryo, Mammalian/physiology , Humans , Mice , Molecular Sequence Data , Trisomy
13.
Mech Dev ; 131: 127-36, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24157522

ABSTRACT

Hedgehog (Hh) signaling plays pivotal roles in morphogenesis of several embryonic tissues, including the primitive gut. In the mouse embryo, Sonic hedgehog (Shh) is expressed in endodermal epithelia from the oral cavity to the intestine, and contributes to cell proliferation in the underlying mesenchyme and subsequent differentiation into the gastrointestinal smooth muscle. Three evolutionary conserved non-coding sequences in the region upstream of the Shh coding sequence contain endoderm-specific enhancers for Shh expression. Although Shh expression in the endodermal epithelial lining is mostly attributed to these three enhancers, none of them regulates gene expression in the gastroesophageal epithelium. Here, we found that a 1.7Kb fragment located 100Kb upstream of the Shh coding sequence contains a functional element for Shh expression in endodermal organs, including the esophagus and stomach. Compared with the three known endodermal enhancers, this novel enhancer shows less evolutionary conservation, even among rodents. In mouse embryonic endodermal tissues, the seamless expression of Shh is achieved by a patchwork of multiple enhancers with different rates of evolution.


Subject(s)
Cell Differentiation/genetics , Gastrointestinal Tract/growth & development , Hedgehog Proteins/biosynthesis , Lung/growth & development , Regulatory Elements, Transcriptional/genetics , Animals , Embryo, Mammalian/metabolism , Endoderm/growth & development , Endoderm/metabolism , Gastrointestinal Tract/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Lung/metabolism , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Morphogenesis , Signal Transduction
14.
Genome Res ; 23(10): 1740-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23878157

ABSTRACT

Coelacanths are known as "living fossils," as they show remarkable morphological resemblance to the fossil record and belong to the most primitive lineage of living Sarcopterygii (lobe-finned fishes and tetrapods). Coelacanths may be key to elucidating the tempo and mode of evolution from fish to tetrapods. Here, we report the genome sequences of five coelacanths, including four Latimeria chalumnae individuals (three specimens from Tanzania and one from Comoros) and one L. menadoensis individual from Indonesia. These sequences cover two African breeding populations and two known extant coelacanth species. The genome is ∼2.74 Gbp and contains a high proportion (∼60%) of repetitive elements. The genetic diversity among the individuals was extremely low, suggesting a small population size and/or a slow rate of evolution. We found a substantial number of genes that encode olfactory and pheromone receptors with features characteristic of tetrapod receptors for the detection of airborne ligands. We also found that limb enhancers of bmp7 and gli3, both of which are essential for limb formation, are conserved between coelacanth and tetrapods, but not ray-finned fishes. We expect that some tetrapod-like genes may have existed early in the evolution of primitive Sarcopterygii and were later co-opted to adapt to terrestrial environments. These coelacanth genomes will provide a cornerstone for studies to elucidate how ancestral aquatic vertebrates evolved into terrestrial animals.


Subject(s)
Adaptation, Biological , Evolution, Molecular , Fishes/classification , Fishes/genetics , Genome , Africa , Animals , Aquatic Organisms/genetics , Base Sequence , Biodiversity , Bone Morphogenetic Protein 7/genetics , Extremities/growth & development , Genetic Speciation , Genetic Variation , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Phylogeny , Receptors, Odorant/genetics , Receptors, Pheromone/genetics , Sequence Analysis, DNA , Vertebrates/classification , Vertebrates/genetics , Water
15.
Hum Mol Genet ; 22(12): 2471-81, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23449628

ABSTRACT

Partial trisomy distal 4q (denoted 4q+) is a human chromosomal disorder caused by duplication of the distal end of the long arm of chromosome 4 (Chr4). This disorder manifests typical phenotypes, including craniofacial, renal, heart and thumb developmental defects. Although these clinical features are likely caused by a dosage imbalance in the gene network involving the trisomic region, the causative gene or genes and the molecular bases are largely unknown. Here, we report mouse Recombination-induced mutation 4 (Rim4) as a model animal of 4q+. The Rim4 genome contains an insertion of a 6.5 Mb fragment from mouse chromosome 8 into chromosome 6. This insertion fragment contains 17 genes, including Hand2, that encode the basic helix-loop-helix transcription factor and is syntenic to the distal end of human Chr4, 4q32.3 to 4q34.1, which is responsible for 4q+. A comparison of phenotypes between patients with Rim4 and 4q+ revealed that Rim4 shows direct parallels with many phenotypes of 4q+ such as craniofacial, heart, cervical vertebra and limb deformities. Rebalancing the gene dosage by a genetic cross with Hand2 knockout mice ameliorated symptoms of the heart and limb deformities of Rim4. Conversely, an increase in copy number of Hand2 in wild-type mice recaptures the heart and limb deformities of Rim4. Our results collectively demonstrate that overdosage of Hand2 is a major cause for at least the limb and heart phenotypes of 4q+ and that mouse Rim4 provides a unique animal model for understanding the molecular bases underlying the complex phenotypes of 4q+.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Dosage , Heart Defects, Congenital/genetics , Limb Deformities, Congenital/genetics , Trisomy/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromosome Disorders/genetics , Chromosome Disorders/metabolism , Chromosomes, Human, Pair 4/genetics , Disease Models, Animal , Extremities/growth & development , Female , Heart/growth & development , Heart Defects, Congenital/metabolism , Humans , Limb Deformities, Congenital/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
J Invest Dermatol ; 131(12): 2477-85, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21776009

ABSTRACT

Despite a strong clinical need for inducing scarless wound healing, the molecular factors required to accomplish it are unknown. Although skin-wound healing in adult mammals often results in scarring, some amphibians can regenerate injured body parts, even an amputated limb, without it. To understand the mechanisms of perfect skin-wound healing in regenerative tetrapods, we studied the healing process in young adult Xenopus "froglets" after experimental skin excision. We found that the excision wound healed completely in Xenopus froglets, without scarring. Mononuclear cells expressing a homeobox gene, prx1, accumulated under the new epidermis of skin wounds on the limb and trunk and at the regenerating limb. In transgenic Xenopus froglets expressing a reporter for the mouse prx1 limb-specific enhancer, activity was seen in the healing skin and in the regenerating limb. Comparable activity did not accompany skin-wound healing in adult mice. Our results suggest that scarless skin-wound healing may require activation of the prx1 limb enhancer, and competence to activate the enhancer is probably a prerequisite for epimorphic regeneration, such as limb regeneration. Finally, the induction of this prx1 enhancer activity may be useful as a reliable marker for therapeutically induced scarless wound healing in mammals.


Subject(s)
Homeodomain Proteins/genetics , Wound Healing/genetics , Xenopus Proteins/genetics , Animals , Animals, Genetically Modified , Cicatrix/genetics , Extremities/physiology , Gene Expression , Mice , Monocytes/metabolism , Promoter Regions, Genetic , Skin/metabolism , Xenopus laevis
17.
Dev Growth Differ ; 52(2): 223-34, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20100248

ABSTRACT

In tetrapod limbs, an anteriormost digit has common traits of small, short and less-phalange morphology. In this study, we focused on three genes, Mkp3, Sef and Tsukushi (TSK), which have anterior-specific or anterior-prominent expression patterns in the developing limb bud at the autopod-forming stage. The anterior expression is not fixed in the period of limb development, but the expression domains of Mkp3, Sef and TSK change considerably from the distal domain to the anterior domain. This change in expression domains, anterior shift, of these genes involves maintenance of gene expression in the anterior side and downregulation in the posterior side. Manipulated overdose of fibroblast growth factor (FGF) in the presumptive digit 2 region of chick forelimb bud results in elongation of cartilage elements of digit 2, suggesting that attenuated FGF signaling, which Mkp3, Sef, and TSK negatively regulate, provides digit 2-specific traits of morphology. The anterior expression of Mkp3 and Sef but not TSK is conserved also in limb buds of the mouse and gecko, and the anterior shift of these genes, accumulation of their transcripts in the anterior side and appropriate regulation of strength of FGF signaling may control species-specific morphology of the anteriormost digit.


Subject(s)
Dual Specificity Phosphatase 6/genetics , Extremities/anatomy & histology , Extremities/embryology , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/genetics , Protein-Tyrosine Kinases/genetics , Animals , Chick Embryo , Fibroblast Growth Factors/metabolism , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, Inbred Strains , Signal Transduction/genetics
18.
Dev Biol ; 335(2): 396-406, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19766620

ABSTRACT

Developing vertebrate limbs are often utilized as a model for studying pattern formation and morphogenetic cell death. Herein, we report that conditional deletion of Rac1, a member of the Rho family of proteins, in mouse limb bud mesenchyme led to skeletal deformities in the autopod and soft tissue syndactyly, with the latter caused by a complete absence of interdigital programmed cell death. Furthermore, the lack of interdigital programmed cell death and associated syndactyly was related to down-regulated gene expression of Bmp2, Bmp7, Msx1, and Msx2, which are known to promote apoptosis in the interdigital mesenchyme. Our findings from Rac1 conditional mutants indicate crucial roles for Rac1 in limb bud morphogenesis, especially interdigital programmed cell death.


Subject(s)
Apoptosis/physiology , Limb Buds/embryology , Mesoderm/enzymology , rac1 GTP-Binding Protein/physiology , Animals , Base Sequence , DNA Primers , Genetic Markers , Immunohistochemistry , Mice , Mice, Transgenic , Polymerase Chain Reaction , Wnt Proteins/metabolism
19.
Development ; 136(10): 1665-74, 2009 May.
Article in English | MEDLINE | ID: mdl-19369396

ABSTRACT

The sonic hedgehog (Shh) pathway plays indispensable roles in the morphogenesis of mouse epithelial linings of the oral cavity and respiratory and digestive tubes. However, no enhancers that regulate regional Shh expression within the epithelial linings have been identified so far. In this study, comparison of genomic sequences across mammalian species and teleost fishes revealed three novel conserved non-coding sequences (CNCSs) that cluster in a region 600 to 900 kb upstream of the transcriptional start site of the mouse Shh gene. These CNCSs drive regional transgenic lacZ reporter expression in the epithelial lining of the oral cavity, pharynx, lung and gut. Together, these enhancers recapitulate the endogenous Shh expression domain within the major epithelial linings. Notably, genomic arrangement of the three CNCSs shows co-linearity that mirrors the order of the epithelial expression domains along the anteroposterior body axis. The results suggest that the three CNCSs are epithelial lining-specific long-range Shh enhancers, and that their actions partition the continuous epithelial linings into three domains: ectoderm-derived oral cavity, endoderm-derived pharynx, and respiratory and digestive tubes of the mouse. Targeted deletion of the pharyngeal epithelium specific CNCS results in loss of endogenous Shh expression in the pharynx and postnatal lethality owing to hypoplasia of the soft palate, epiglottis and arytenoid. Thus, this long-range enhancer is indispensable for morphogenesis of the pharyngeal apparatus.


Subject(s)
Hedgehog Proteins/biosynthesis , Intestinal Mucosa/metabolism , Mouth Mucosa/metabolism , Respiratory Mucosa/metabolism , Animals , Cell Lineage , Ectoderm/cytology , Ectoderm/embryology , Endoderm/cytology , Endoderm/embryology , Enhancer Elements, Genetic , Epithelium/embryology , Epithelium/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Mice , Mice, Knockout , Mouth Mucosa/cytology , Mouth Mucosa/embryology , Pharynx/cytology , Pharynx/embryology , Pharynx/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/embryology
20.
Dev Cell ; 16(1): 47-57, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19097946

ABSTRACT

The expression of Sonic hedgehog (Shh) in mouse limb buds is regulated by a long-range enhancer 1 Mb upstream of the Shh promoter. We used 3D-FISH and chromosome conformation capture assays to track changes at the Shh locus and found that long-range promoter-enhancer interactions are specific to limb bud tissues competent to express Shh. However, the Shh locus loops out from its chromosome territory only in the posterior limb bud (zone of polarizing activity or ZPA), where Shh expression is active. Notably, while Shh mRNA is detected throughout the ZPA, enhancer-promoter interactions and looping out were only observed in small fractions of ZPA cells. In situ detection of nascent Shh transcripts and unstable EGFP reporters revealed that active Shh transcription is likewise only seen in a small fraction of ZPA cells. These results suggest that chromosome conformation dynamics at the Shh locus allow transient pulses of Shh transcription.


Subject(s)
Chromosomes , Gene Expression Regulation, Developmental , Hedgehog Proteins , Limb Buds/physiology , Transcription, Genetic , Animals , Chromosomes/metabolism , Chromosomes/ultrastructure , Enhancer Elements, Genetic , Genes, Reporter , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , In Situ Hybridization, Fluorescence , Limb Buds/anatomy & histology , Mice , Mice, Inbred C57BL , Morphogenesis , RNA Precursors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...