Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biomed Res Int ; 2021: 4084371, 2021.
Article in English | MEDLINE | ID: mdl-33553423

ABSTRACT

OBJECTIVE: Sepsis is a leading cause of acute lung injury (ALI). This study attempted to investigate the effects of limb bud and heart (LBH) on the development of sepsis-induced ALI and its underlying mechanism of action. METHODS: The sepsis-induced ALI mouse model was established by cecal ligation and puncture (CLP). The lung injury score and lung wet/dry weight (W/D) ratio were used to evaluate the lung injury. In vitro, ALI was simulated by lipopolysaccharide (LPS) treatment in A549 cells. The mRNA expression of LBH, NLRP3, ASC, and proinflammatory cytokines was measured by qRT-PCR. The viability of LPS-induced A549 cells was analyzed by MTT assay. Furthermore, western blot was performed to detect the protein expression of LBH, NLRP3, and ASC. LPS-induced A549 cells were treated with MCC950 (NLRP3 inflammasome inhibitor) to confirm the effect of LBH on NLRP3 inflammasome. RESULTS: The mRNA and protein expression of LBH was decreased in sepsis-induced ALI. LBH overexpression reduced the lung injury score, lung W/D ratio, expression of proinflammatory cytokines, and NLRP3 inflammasome activation in sepsis-induced ALI mouse model. Additionally, LBH upregulation increased the viability, while it decreased the proinflammatory cytokine expression and NLRP3 inflammasome activation of LPS-induced A549 cells. Moreover, MCC950 reversed the promoting effects of LBH silencing on proinflammatory cytokine expression and NLRP3 inflammasome activation in LPS-induced A549 cells. CONCLUSIONS: LBH alleviated lung injury in sepsis-induced ALI mouse model by inhibiting inflammation and NLRP3 inflammasome, and restrained the inflammation by inhibiting NLRP3 inflammasome in LPS-induced A549 cells, providing a novel therapeutic target for ALI.


Subject(s)
Acute Lung Injury/genetics , CARD Signaling Adaptor Proteins/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Sepsis/genetics , Transcription Factors/genetics , A549 Cells , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Gene Expression Regulation/drug effects , Heart/physiopathology , Humans , Inflammasomes/genetics , Limb Buds/metabolism , Limb Buds/pathology , Lipopolysaccharides/pharmacology , Mice , Sepsis/chemically induced , Sepsis/complications , Sepsis/pathology
2.
Genet Med ; 22(1): 189-198, 2020 01.
Article in English | MEDLINE | ID: mdl-31395945

ABSTRACT

PURPOSE: Preaxial polydactyly (PPD) is a common congenital hand malformation classified into four subtypes (PPD I-IV). Variants in the zone of polarizing activity regulatory sequence (ZRS) within intron 5 of the LMBR1 gene are linked to most PPD types. However, the genes responsible for PPD I and the underlying mechanisms are unknown. METHODS: A rare large four-generation family with isolated PPD I was subjected to genome-wide genotyping and sequence analysis. In vitro and in vivo functional studies were performed in Caco-2 cells, 293T cells, and a knockin transgenic mouse model. RESULTS: A novel g.101779T>A (reference sequence: NG_009240.2; position 446 of the ZRS) variant segregates with all PPD I-affected individuals. The knockin mouse with this ZRS variant exhibited PPD I phenotype accompanying ectopic and excess expression of Shh. We confirmed that HnRNP K can bind the ZRS and SHH promoters. The ZRS mutant enhanced the binding affinity for HnRNP K and upregulated SHH expression. CONCLUSION: Our results identify the first PPD I disease-causing variant. The variant leading to PPD I may be associated with enhancing SHH expression mediated by HnRNP K. This study adds to the ZRS-associated syndromes classification system for PPD and clarifies the underlying molecular mechanisms.


Subject(s)
Hedgehog Proteins/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Limb Buds/growth & development , Membrane Proteins/genetics , Polydactyly/genetics , Polymorphism, Single Nucleotide , Thumb/abnormalities , Up-Regulation , Animals , Caco-2 Cells , Disease Models, Animal , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Introns , Limb Buds/metabolism , Limb Buds/pathology , Male , Mice , Mice, Transgenic , Pedigree , Polydactyly/metabolism
3.
Bone ; 105: 57-66, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28826843

ABSTRACT

Tyrosine kinase inhibitors are being developed for therapy of malignancies caused by oncogenic FGFR signaling but little is known about their effect in congenital chondrodysplasias or craniosynostoses that associate with activating FGFR mutations. Here, we investigated the effects of novel FGFR inhibitor, ARQ 087, in experimental models of aberrant FGFR3 signaling in cartilage. In cultured chondrocytes, ARQ 087 efficiently rescued all major effects of pathological FGFR3 activation, i.e. inhibition of chondrocyte proliferation, loss of extracellular matrix and induction of premature senescence. In ex vivo tibia organ cultures, ARQ 087 restored normal growth plate architecture and eliminated the suppressing FGFR3 effect on chondrocyte hypertrophic differentiation, suggesting that it targets the FGFR3 pathway specifically, i.e. without interference with other pro-growth pathways. Moreover, ARQ 087 inhibited activity of FGFR1 and FGFR2 mutants associated with Pfeiffer, Apert and Beare-Stevenson craniosynostoses, and rescued FGFR-driven excessive osteogenic differentiation in mouse mesenchymal micromass cultures or in ex vivo calvarial organ cultures. Our data warrant further development of ARQ 087 for clinical use in skeletal disorders caused by activating FGFR mutations.


Subject(s)
Aniline Compounds/therapeutic use , Cell Differentiation , Chondrocytes/pathology , Craniosynostoses/drug therapy , Craniosynostoses/pathology , Mutation/genetics , Quinazolines/therapeutic use , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction , Aniline Compounds/pharmacology , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell-Free System , Cellular Senescence/drug effects , Chickens , Chondrocytes/drug effects , Chondrocytes/metabolism , Craniosynostoses/genetics , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblast Growth Factor 2/pharmacology , Limb Buds/pathology , Mice , Organ Culture Techniques , Quinazolines/pharmacology , Rats , Skull/pathology , Tibia/drug effects , Tibia/pathology
4.
Dev Biol ; 424(1): 1-9, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28235582

ABSTRACT

Matching appendage size to body size is fundamental to animal function. Generating an appropriately-sized appendage is a robust process executed during development which is also critical for regeneration. When challenged, larger animals are programmed to regenerate larger limbs than smaller animals within a single species. Understanding this process has important implications for regenerative medicine. To approach this complex question, models with altered appendage size:body size ratios are required. We hypothesized that repeatedly challenging axolotls to regrow limb buds would affect their developmental program resulting in altered target morphology. We discovered that after 10 months following this experimental procedure, limbs that developed were permanently miniaturized. This altered target morphology was preserved upon amputation and regeneration. Future experiments using this platform should provide critical information about how target limb size is encoded within limb progenitors.


Subject(s)
Ambystoma mexicanum/embryology , Amputation, Surgical , Limb Buds/embryology , Limb Buds/pathology , Animals , Ectromelia/pathology , Limb Buds/abnormalities , Limb Buds/innervation , Nerve Tissue/pathology , Organ Size , Regeneration
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.
Hum Mol Genet ; 21(18): 3941-55, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22634226

ABSTRACT

Achondroplasia (ACH) and thanatophoric dysplasia (TD) are caused by gain-of-function mutations of fibroblast growth factor receptor 3 (FGFR3) and they are the most common forms of dwarfism and lethal dwarfism, respectively. Currently, there are few effective treatments for ACH. For the neonatal lethality of TD patients, no practical effective therapies are available. We here showed that systemic intermittent PTH (1-34) injection can rescue the lethal phenotype of TD type II (TDII) mice and significantly alleviate the retarded skeleton development of ACH mice. PTH-treated ACH mice had longer naso-anal length than ACH control mice, and the bone lengths of humeri and tibiae were rescued to be comparable with those of wild-type control mice. Our study also found that the premature fusion of cranial synchondroses in ACH mice was partially corrected after the PTH (1-34) treatment, suggesting that the PTH treatment may rescue the progressive narrowing of neurocentral synchondroses that cannot be readily corrected by surgery. In addition, we found that the PTH treatment can improve the osteopenia and bone structure of ACH mice. The increased expression of PTHrP and down-regulated FGFR3 level may be responsible for the positive effects of PTH on bone phenotype of ACH and TDII mice.


Subject(s)
Achondroplasia/drug therapy , Bone Density Conservation Agents/administration & dosage , Bone Development/drug effects , Teriparatide/administration & dosage , Thanatophoric Dysplasia/drug therapy , Achondroplasia/genetics , Achondroplasia/pathology , Animals , Body Weight/drug effects , Bone Density/drug effects , Bone Density Conservation Agents/pharmacology , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/genetics , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Bone and Bones/pathology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/physiology , Drug Evaluation, Preclinical , Gene Expression , Gene Expression Regulation , Humans , Limb Buds/drug effects , Limb Buds/pathology , Mice , Mice, Transgenic , Mutation, Missense , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Radiography , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Teriparatide/pharmacology , Thanatophoric Dysplasia/genetics , Thanatophoric Dysplasia/pathology , Tissue Culture Techniques
7.
J Bone Miner Metab ; 30(3): 270-80, 2012 May.
Article in English | MEDLINE | ID: mdl-21960178

ABSTRACT

Maternal malnutrition during pregnancy was shown by numerous studies to result in the birth of offspring exhibiting altered bone characteristics, which are indicative of bone loss. We hypothesized that not only maternal malnutrition but also some developmental toxicants (teratogens) given at a dose inducing neither structural anomalies nor growth retardation can detrimentally affect skeletal health in adult offspring. To check this hypothesis, pregnant mice were exposed to a single injection of 5-aza-2-deoxycytidine (5-AZA) (a teratogen capable of inducing phocomelia of the hind limbs) at a sub-threshold teratogenic dose. Micro-computed tomography scanning revealed that femora of 5-month-old male offspring exposed in uterus to 5-AZA had trabecular microarchitecture indicative of bone loss. Furthermore, exposure to 5-AZA increased the susceptibility of offspring to postnatal chronic mild stress, which has been shown to induce bone loss in mice. While exploring possible mechanisms underlying this phenomenon, we observed that the expression of some microRNAs, which have been demonstrated as regulators of key osteoblastogenic genes, was altered in hind limb buds of embryos exposed to 5-AZA. Furthermore, the expression of receptor activator of nuclear factor kappa B ligand (RANKL) in femoral stromal/osteoblastic cells of 5-month-old offspring of 5-AZA-treated females was found to be increased. Collectively, this study implies for the first time that single low-dose exposure to a teratogen can induce bone loss in adult offspring, possibly via alteration of embryonic microRNAs and RANKL expression.


Subject(s)
Aging/drug effects , Aging/pathology , Azacitidine/toxicity , Bone Resorption/chemically induced , Prenatal Exposure Delayed Effects/pathology , Teratogens/toxicity , Acid Phosphatase/metabolism , Animals , Apoptosis/drug effects , Bone Resorption/diagnostic imaging , Bone Resorption/genetics , Bone Resorption/pathology , Embryo, Mammalian/drug effects , Embryo, Mammalian/pathology , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Hindlimb/drug effects , Hindlimb/embryology , Hindlimb/metabolism , Hindlimb/pathology , Isoenzymes/metabolism , Limb Buds/drug effects , Limb Buds/metabolism , Limb Buds/pathology , Mice , Mice, Inbred ICR , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Osteogenesis/genetics , Osteoprotegerin/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/genetics , RANK Ligand/metabolism , Reproduction/drug effects , Tartrate-Resistant Acid Phosphatase , X-Ray Microtomography
8.
PLoS One ; 6(11): e27741, 2011.
Article in English | MEDLINE | ID: mdl-22140465

ABSTRACT

Xenografting is widely used for assessing in vivo pluripotency of human stem cell populations. Here, we report on early to late events in the development of mature experimental teratoma from a well-characterized human embryonic stem cell (HESC) line, HS181. The results show an embryonic process, increasingly chaotic. Active proliferation of the stem cell derived cellular progeny was detected already at day 5, and characterized by the appearance of multiple sites of engraftment, with structures of single or pseudostratified columnar epithelium surrounding small cavities. The striking histological resemblance to developing embryonic ectoderm, and the formation of epiblast-like structures was supported by the expression of the markers OCT4, NANOG, SSEA-4 and KLF4, but a lack of REX1. The early neural marker NESTIN was uniformly expressed, while markers linked to gastrulation, such as BMP-4, NODAL or BRACHYURY were not detected. Thus, observations on day 5 indicated differentiation comparable to the most early transient cell populations in human post implantation development. Confirming and expanding on previous findings from HS181 xenografts, these early events were followed by an increasingly chaotic development, incorporated in the formation of a benign teratoma with complex embryonic components. In the mature HS181 teratomas not all types of organs/tissues were detected, indicating a restricted differentiation, and a lack of adequate spatial developmental cues during the further teratoma formation. Uniquely, a kinetic alignment of rare complex structures was made to human embryos at diagnosed gestation stages, showing minor kinetic deviations between HS181 teratoma and the human counterpart.


Subject(s)
Embryonic Stem Cells/cytology , Germ Layers/cytology , Transplantation, Heterologous , Biomarkers/metabolism , Cell Differentiation , Cell Line , Embryonic Stem Cells/metabolism , Germ Layers/metabolism , Humans , Kruppel-Like Factor 4 , Limb Buds/metabolism , Limb Buds/pathology , Neurons/metabolism , Neurons/pathology , Teratoma/metabolism , Teratoma/pathology , Time Factors
9.
Bone ; 49(4): 673-82, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21807129

ABSTRACT

RUNX2 is an essential transcription factor for osteoblast differentiation, because osteoblast differentiation is completely blocked in Runx2-deficient mice. However, it remains to be clarified whether RUNX2 is sufficient for osteoblast differentiation during embryogenesis. To address this issue, Runx2 transgenic mice were generated under the control of the Prrx1 promoter, which directs the transgene expression to mesenchymal cells before the onset of bone development. The transgene expression was detected in the cranium, limb buds, and the region from the mandible to anterior chest wall. The skull became small and the limbs were shortened depending on the levels of the transgene expression. Early onset of Runx2 expression in the cranial mesenchyme induced mineralization on E13.0, when no mineralization was observed in wild-type mice, and resulted in craniosynostosis as shown by the closure of sutures and fontanelles on E18.5. Col1a1 and Spp1 expressions were detected in the mineralized regions on E12.5-13.5. The limb bones were hypoplastic and fused, and ectopic bones were formed in the hands and feet. Col2a1 expression was inhibited but Col1a1 expression was induced in the limb buds on E12.5. In the anterior chest wall, ectopic bones were formed through the process of intramembranous ossification, interrupting the formation of cartilaginous anlagen of sternal manubrium. These findings indicate that RUNX2 is sufficient to direct mesenchymal cells to osteoblasts and lead to intramembranous bone formation during embryogenesis; Runx2 inhibits chondrocyte differentiation at an early stage; and that Runx2 expression at appropriate level, times and spaces during embryogenesis is essential for skeletal development.


Subject(s)
Choristoma/complications , Choristoma/embryology , Core Binding Factor Alpha 1 Subunit/metabolism , Craniosynostoses/complications , Craniosynostoses/embryology , Limb Buds/abnormalities , Osteogenesis , Animals , Bone and Bones/diagnostic imaging , Bone and Bones/embryology , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/metabolism , Cartilage/pathology , Cell Differentiation , Chondrocytes/metabolism , Chondrocytes/pathology , Choristoma/diagnostic imaging , Choristoma/pathology , Craniosynostoses/diagnostic imaging , Craniosynostoses/pathology , Face , Fluorescence , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Limb Buds/diagnostic imaging , Limb Buds/metabolism , Limb Buds/pathology , Mice , Mice, Transgenic , Osteoblasts/metabolism , Osteoblasts/pathology , Skull/diagnostic imaging , Skull/embryology , Skull/pathology , X-Ray Microtomography
11.
Carcinogenesis ; 30(10): 1789-95, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19635748

ABSTRACT

Mice bearing germ line mutations of p53 develop sarcomas at a significant rate. Since they are susceptible to a variety of other malignancies, they are not ideally suited to the study of sarcomas. To test the possibility that targeted mutation of tumor suppressor genes in early mesenchymal cells would induce formation of sarcomas, the Prx1-cre transgenic mouse was crossed to mice-bearing floxed alleles of p53 and Rb. Mice with homozygous deletion of p53 (Prx1-cre p53(lox/lox)) developed sarcomas in the extremities at a mean time of 50 weeks. Osteosarcomas (OS) were the most common type of sarcoma (61%) followed by poorly differentiated soft tissue sarcomas (PDSTS) (32%). Homozygous deletion of p53 produced sarcomas significantly more rapidly than heterozygous deletion, which resulted in sarcoma formation after a mean of 96 weeks. Mice with homozygous Rb mutation (Prx1-cre Rb(lox/lox)) developed normally and had no ostensible defects in the limbs. In contrast to p53, targeted deletion of Rb did not produce sarcomas in the limbs. However, simultaneous deletion of Rb and p53 accelerated the time to sarcoma formation, and a greater percentage of PDSTS were found. Deletion of p53 in committed osteoblasts by the Col1a1-cre transgenic mouse bearing an osteoblast-specific enhancer resulted in a high percentage of OS. These findings suggest that deletion of p53 in mesenchymal cells that give rise to osteoblasts is a powerful initiator of OS. Deletion of Rb does not initiate sarcoma formation in mice, but it accelerates formation of both soft tissue sarcomas and OS.


Subject(s)
Genes, Retinoblastoma , Genes, p53 , Limb Buds/pathology , Osteosarcoma/genetics , Sarcoma, Experimental/genetics , Animals , Crosses, Genetic , Gene Deletion , Heterozygote , Homozygote , Mice , Mutation , Osteosarcoma/pathology , Promoter Regions, Genetic , Sarcoma, Experimental/pathology
12.
Nature ; 460(7253): 400-4, 2009 Jul 16.
Article in English | MEDLINE | ID: mdl-19553938

ABSTRACT

Phocomelia is a devastating, rare congenital limb malformation in which the long bones are shorter than normal, with the upper portion of the limb being most severely affected. In extreme cases, the hands or fingers are attached directly to the shoulder and the most proximal elements (those closest to the shoulder) are entirely missing. This disorder, previously known in both autosomal recessive and sporadic forms, showed a marked increase in incidence in the early 1960s due to the tragic toxicological effects of the drug thalidomide, which had been prescribed as a mild sedative. This human birth defect is mimicked in developing chick limb buds exposed to X-irradiation. Both X-irradiation and thalidomide-induced phocomelia have been interpreted as patterning defects in the context of the progress zone model, which states that a cell's proximodistal identity is determined by the length of time spent in a distal limb region termed the 'progress zone'. Indeed, studies of X-irradiation-induced phocomelia have served as one of the two major experimental lines of evidence supporting the validity of the progress zone model. Here, using a combination of molecular analysis and lineage tracing in chick, we show that X-irradiation-induced phocomelia is fundamentally not a patterning defect, but rather results from a time-dependent loss of skeletal progenitors. Because skeletal condensation proceeds from the shoulder to fingers (in a proximal to distal direction), the proximal elements are differentially affected in limb buds exposed to radiation at early stages. This conclusion changes the framework for considering the effect of thalidomide and other forms of phocomelia, suggesting the possibility that the aetiology lies not in a defect in the patterning process, but rather in progenitor cell survival and differentiation. Moreover, molecular evidence that proximodistal patterning is unaffected after X-irradiation does not support the predictions of the progress zone model.


Subject(s)
Body Patterning/radiation effects , Ectromelia/etiology , Ectromelia/pathology , Limb Buds/pathology , Limb Buds/radiation effects , Animals , Bone and Bones/cytology , Bone and Bones/radiation effects , Cell Death/radiation effects , Cell Differentiation/radiation effects , Cell Lineage/radiation effects , Cell Proliferation/radiation effects , Chick Embryo , Chondrogenesis/radiation effects , Ectromelia/genetics , Gene Expression Regulation, Developmental/radiation effects , Limb Buds/abnormalities , Limb Buds/transplantation , Reproducibility of Results , Stem Cells/cytology , Stem Cells/radiation effects , Thalidomide/adverse effects , Time Factors , X-Rays/adverse effects
13.
Handchir Mikrochir Plast Chir ; 36(2-3): 108-16, 2004.
Article in English | MEDLINE | ID: mdl-15162308

ABSTRACT

In order to better understand the teratogenic mechanisms of congenital defects of the digits, we analyzed clinical cases and induced similar types of congenital hand anomalies in rat fetuses by oral administration of busulfan. In clinical cases, radial and ulnar deficiencies had common characteristic features. We induced radial and ulnar deficiencies in rat fetuses with the same drug. Radial and ulnar deficiencies induced in rats have similar clinical manifestations and these anomalies might be caused by the same teratogenic mechanism. Then, the formation of the digital rays was examined histologically. The results of histological examination suggested that these deficiencies were not caused by localized damage of the limb bud. They also suggested that the cause of missing digits in longitudinal deficiency is closely related to a deficit of mesenchymal cells in the limb bud. Cleft hand is considered to be one of the types of longitudinal deficiency. However, several investigators have suggested that the abnormal induction of finger rays in the process of formation of fingers induced central polydactyly, osseous syndactyly and also cleft hand. X-rays of the clinical cases and skeletal changes of the anomalies induced in rats appear to demonstrate that cleft hand formation proceeds from osseous syndactyly and central polydactyly. The results of our experimental study show that the critical periods of central polydactyly, osseous syndactyly and cleft hand are the same. They also suggest that central polydactyly, syndactyly and cleft hand might be induced when the same teratogenic factor acts on embryos at the same developmental stage in the human being. Because they have a similar causation, cleft hand, syndactyly and central polydactyly should be classified into the same entity, that is, abnormal induction of digital rays. Based on these clinical and experimental studies, we modified the Swanson classification. In our modified classification, typical cleft hand, syndactyly and polydactyly are included in the same category of abnormal induction of digital rays as the fourth new category.


Subject(s)
Ectromelia/chemically induced , Hand Deformities, Congenital/chemically induced , Teratogens , Animals , Busulfan , Ectromelia/embryology , Ectromelia/pathology , Female , Fingers/abnormalities , Fingers/pathology , Gestational Age , Hand Deformities, Congenital/embryology , Hand Deformities, Congenital/pathology , Humans , Infant , Infant, Newborn , Limb Buds/drug effects , Limb Buds/embryology , Limb Buds/pathology , Polydactyly/chemically induced , Polydactyly/embryology , Polydactyly/pathology , Pregnancy , Radius/abnormalities , Radius/embryology , Radius/pathology , Rats , Rats, Inbred Strains , Syndactyly/chemically induced , Syndactyly/embryology , Syndactyly/pathology , Ulna/abnormalities , Ulna/embryology , Ulna/pathology
14.
Radiat Res ; 154(6): 673-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11096424

ABSTRACT

We reported previously that in utero radiation-induced apoptosis in the predigital regions of embryonic limb buds was responsible for digital defects in mice. To investigate the possible involvement of the Trp53 gene, the present study was conducted using embryonic C57BL/6J mice with different Trp53 status. Susceptibility to radiation-induced apoptosis in the predigital regions and digital defects depended on both Trp53 status and the radiation dose; i.e., Trp53 wild-type (Trp53(+/+)) mice appeared to be the most sensitive, Trp53 heterozygous (Trp53(+/-)) mice were intermediate, and Trp53 knockout (Trp53(-/-)) mice were the most resistant. These results indicate that induction of apoptosis and digital defects by prenatal irradiation in the later period of organogenesis are mediated by the Trp53 gene. These findings suggest that the wild-type Trp53 gene may be an intrinsic genetic susceptibility factor that is responsible for certain congenital defects induced by prenatal irradiation.


Subject(s)
Abnormalities, Radiation-Induced/genetics , Apoptosis/radiation effects , Limb Deformities, Congenital , Prenatal Exposure Delayed Effects , Tumor Suppressor Protein p53/genetics , Abnormalities, Radiation-Induced/pathology , Animals , Dose-Response Relationship, Radiation , Female , Fetal Death/pathology , Limb Buds/pathology , Limb Buds/radiation effects , Limb Deformities, Congenital/diagnostic imaging , Limb Deformities, Congenital/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Radiation Tolerance/genetics , Radiography , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism
15.
Mech Dev ; 81(1-2): 115-25, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10330489

ABSTRACT

We describe here how the early limb bud of the quail embryo develops in the absence of retinoids, including retinoic acid. Retinoid-deficient embryos develop to about stage 20/21, thus allowing patterns of early gene activity in the limb bud to be readily examined. Genes representing different aspects of limb polarity were analysed. Concerning the anteroposterior axis, Hoxb-8 was up-regulated and its border was shifted anteriorly whereas shh and the mesodermal expression of bmp-2 were down-regulated in the absence of retinoids. Concerning the apical ectodermal genes, fgf-4 was down-regulated whereas fgf-8 and the ectodermal domain of bmp-2 were unaffected. Genes involved in dorsoventral polarity were all disrupted. Wnt-7a, normally confined to the dorsal ectoderm, was ectopically expressed in the ventral ectoderm and the corresponding dorsal mesodermal gene Lmx-1 spread into the ventral mesoderm. En-1 was partially or completely absent from the ventral ectoderm. These dorsoventral patterns of expression resemble those seen in En-1 knockout mouse limb buds. Overall, the patterns of gene expression are also similar to the Japanese limbless mutant. These experiments demonstrate that the retinoid-deficient embryo is a valuable tool for dissecting pathways of gene activity in the limb bud and reveal for the first time a role for retinoic acid in the organisation of the dorsoventral axis.


Subject(s)
Body Patterning , Coturnix/embryology , Limb Buds/embryology , Ovum/metabolism , Retinoids/metabolism , Trans-Activators , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Fibroblast Growth Factor 4 , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins , Homeodomain Proteins/metabolism , In Situ Hybridization , LIM-Homeodomain Proteins , Limb Buds/pathology , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors , Wnt Proteins
16.
Teratology ; 56(4): 262-70, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9408977

ABSTRACT

Dactylaplasia, or Dac, was recently mapped to the distal portion of mouse chromosome 19 and shown to be inherited as an autosomal semi-dominant trait characterized by missing central digital rays. The most common locus for human split hand split foot malformation, also typically characterized by missing central digital rays, is 10q25, a region of synteny to the Dac locus. The Dac mouse appears to be an ideal genotypic and phenotypic model for this human malformation syndrome. Several genes lie in this region of synteny, however, only Fibroblast Growth Factor 8, or Fgf-8, has been implicated to have a role in limb development. We demonstrate that the developmental mechanism underlying loss of central rays in Dac limbs is dramatic cell death of the apical ectodermal ridge, or AER. This cell death pattern is apparent in E10.5-11.5 Dac limb buds stained with the supravital dye Nile Blue Sulfate. We demonstrate that Fgf8 expression in wild type limbs colocalizes spatially and temporally with AER cell death in Dac limbs. Furthermore, in our mapping panel, there is an absence of recombinants between Fgf-8 and the Dac locus in 133 backcross progeny with a median linkage estimate of approximately 0.5 cM. Thus, our results demonstrate that cell death of the AER in Dac limbs silences the role of the AER as key regulator of limb outgrowth, and that Fgf-8 is a strong candidate for the cause of the Dac phenotype.


Subject(s)
Apoptosis , Ectoderm/pathology , Fibroblast Growth Factors , Limb Buds/abnormalities , Limb Deformities, Congenital/etiology , Animals , Blotting, Northern , Cell Death , Ectoderm/metabolism , Electrophoresis, Polyacrylamide Gel , Female , Fibroblast Growth Factor 8 , Gene Expression Regulation, Developmental , Growth Substances/metabolism , Limb Buds/metabolism , Limb Buds/pathology , Limb Deformities, Congenital/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NZB , Mice, Mutant Strains , Polymerase Chain Reaction , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...