Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Stem Cell Res Ther ; 11(1): 453, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33109263

ABSTRACT

Primary immunodeficiency diseases (PIDs) are rare diseases that are characterized by genetic mutations that damage immunological function, defense, or both. Some of these rare diseases are caused by aberrations in the normal development of natural killer cells (NKs) or affect their lytic synapse. The pathogenesis of these types of diseases as well as the processes underlying target recognition by human NK cells is not well understood. Utilizing induced pluripotent stem cells (iPSCs) will aid in the study of human disorders, especially in the PIDs with defects in NK cells for PID disease modeling. This, together with genome editing technology, makes it possible for us to facilitate the discovery of future therapeutics and/or cell therapy treatments for these patients, because, to date, the only curative treatment available in the most severe cases is hematopoietic stem cell transplantation (HSCT). Recent progress in gene editing technology using CRISPR/Cas9 has significantly increased our capability to precisely modify target sites in the human genome. Among the many tools available for us to study human PIDs, disease- and patient-specific iPSCs together with gene editing offer unique and exceptional methodologies to gain deeper and more thorough understanding of these diseases as well as develop possible alternative treatment strategies. In this review, we will discuss some immunodeficiency disorders affecting NK cell function, such as classical NK deficiencies (CNKD), functional NK deficiencies (FNKD), and PIDs with involving NK cells as well as strategies to model and correct these diseases for further study and possible avenues for future therapies.


Subject(s)
Induced Pluripotent Stem Cells , Primary Immunodeficiency Diseases , Gene Editing , Humans , Killer Cells, Natural , Stem Cell Transplantation
2.
Reprod Domest Anim ; 51 Suppl 2: 18-24, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27762052

ABSTRACT

More than eighteen years have passed since the first derivation of human embryonic stem cells (ESCs), but their clinical use is still met with several challenges, such as ethical concerns regarding the need of human embryos, tissue rejection after transplantation and tumour formation. The generation of human induced pluripotent stem cells (iPSCs) enables the access to patient-derived pluripotent stem cells (PSCs) and opens the door for personalized medicine as tissues/organs can potentially be generated from the same genetic background as the patient recipients, thus avoiding immune rejections or complication of immunosuppression strategies. In this regard, successful replacement, or augmentation, of the function of damaged tissue by patient-derived differentiated stem cells provides a promising cell replacement therapy for many devastating human diseases. Although human iPSCs can proliferate unlimitedly in culture and harbour the potential to generate all cell types in the adult body, currently, the functionality of differentiated cells is limited. An alternative strategy to realize the full potential of human iPSC for regenerative medicine is the in vivo tissue generation in large animal species via interspecies blastocyst complementation. As this technology is still in its infancy and there remains more questions than answers, thus in this review, we mainly focus the discussion on the conceptual framework, the emerging technologies and recent advances involved with interspecies blastocyst complementation, and will refer the readers to other more in-depth reviews on dynamic pluripotent stem cell states, genome editing and interspecies chimeras. Likewise, other emerging alternatives to combat the growing shortage of human organs, such as xenotransplantation or tissue engineering, topics that has been extensively reviewed, will not be covered here.


Subject(s)
Blastocyst/physiology , Induced Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Regenerative Medicine/methods , Animals , Blastocyst/cytology , Cell Differentiation , Cell Proliferation , Gene Editing , Humans , Organ Culture Techniques , Precision Medicine , Sus scrofa
3.
Stem Cells ; 34(9): 2418-28, 2016 09.
Article in English | MEDLINE | ID: mdl-27300161

ABSTRACT

Epigenetic reprogramming is a central process during mammalian germline development. Genome-wide DNA demethylation in primordial germ cells (PGCs) is a prerequisite for the erasure of epigenetic memory, preventing the transmission of epimutations to the next generation. Apart from DNA demethylation, germline reprogramming has been shown to entail reprogramming of histone marks and chromatin remodelling. Contrary to other animal models, there is limited information about the epigenetic dynamics during early germ cell development in humans. Here, we provide further characterization of the epigenetic configuration of the early human gonadal PGCs. We show that early gonadal human PGCs are DNA hypomethylated and their chromatin is characterized by low H3K9me2 and high H3K27me3 marks. Similarly to previous observations in mice, human gonadal PGCs undergo dynamic chromatin changes concomitant with the erasure of genomic imprints. Interestingly, and contrary to mouse early germ cells, expression of BLIMP1/PRDM1 persists in through all gestational stages in human gonadal PGCs and is associated with nuclear lysine-specific demethylase-1. Our work provides important additional information regarding the chromatin changes associated with human PGCs development between 6 and 13 weeks of gestation in male and female gonads. Stem Cells 2016;34:2418-2428.


Subject(s)
Cellular Reprogramming/genetics , Epigenesis, Genetic , Germ Cells/cytology , Germ Cells/metabolism , Gonads/cytology , Animals , Chromatin/metabolism , DNA Methylation/genetics , Female , Histone Demethylases/metabolism , Histones/metabolism , Humans , Lysine/metabolism , Mice , Models, Biological , Positive Regulatory Domain I-Binding Factor 1/metabolism , Species Specificity , Transcription Factors/metabolism
4.
Mol Psychiatry ; 21(9): 1215-24, 2016 09.
Article in English | MEDLINE | ID: mdl-26239292

ABSTRACT

With an onset under the age of 3 years, autism spectrum disorders (ASDs) are now understood as diseases arising from pre- and/or early postnatal brain developmental anomalies and/or early brain insults. To unveil the molecular mechanisms taking place during the misshaping of the developing brain, we chose to study cells that are representative of the very early stages of ontogenesis, namely stem cells. Here we report on MOlybdenum COfactor Sulfurase (MOCOS), an enzyme involved in purine metabolism, as a newly identified player in ASD. We found in adult nasal olfactory stem cells of 11 adults with ASD that MOCOS is downregulated in most of them when compared with 11 age- and gender-matched control adults without any neuropsychiatric disorders. Genetic approaches using in vivo and in vitro engineered models converge to indicate that altered expression of MOCOS results in neurotransmission and synaptic defects. Furthermore, we found that MOCOS misexpression induces increased oxidative-stress sensitivity. Our results demonstrate that altered MOCOS expression is likely to have an impact on neurodevelopment and neurotransmission, and may explain comorbid conditions, including gastrointestinal disorders. We anticipate our discovery to be a fresh starting point for the study on the roles of MOCOS in brain development and its functional implications in ASD clinical symptoms. Moreover, our study suggests the possible development of new diagnostic tests based on MOCOS expression, and paves the way for drug screening targeting MOCOS and/or the purine metabolism to ultimately develop novel treatments in ASD.


Subject(s)
Autism Spectrum Disorder/metabolism , Stem Cells/metabolism , Sulfurtransferases/metabolism , Adult , Animals , Autism Spectrum Disorder/genetics , Caenorhabditis elegans , Female , France , Humans , Male , Mice , Mice, Inbred C57BL , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/physiology , Stem Cells/physiology , Sulfurtransferases/therapeutic use
5.
Sci Rep ; 5: 16400, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26563344

ABSTRACT

Autologous chondrocyte implantation (ACI) depends on the quality and quantity of implanted cells and is hindered by the fact that chondrocytes cultured for long periods of time undergo dedifferentiation. Here we have developed a reproducible and efficient chondrogenic protocol to redifferentiate chondrocytes isolated from osteoarthritis (OA) patients. We used morphological, histological and immunological analysis together with a RT-PCR detection of collagen I and collagen II gene expression to show that chondrocytes isolated from articular cartilage biopsies of patients and subjected to long-term culture undergo dedifferentiation and that these cells can be redifferentiated following treatment with the chimeric Activin A/BMP2 ligand AB235. Examination of AB235-treated cell pellets in both in vitro and in vivo experiments revealed that redifferentiated chondrocytes synthesized a cartilage-specific extracellular matrix (ECM), primarily consisting of vertically-orientated collagen fibres and cartilage-specific proteoglycans. AB235-treated cell pellets also integrated into the surrounding subcutaneous tissue following transplantation in mice as demonstrated by their dramatic increase in size while non-treated control pellets disintegrated upon transplantation. Thus, our findings describe an effective protocol for the promotion of redifferentiation of autologous chondrocytes obtained from OA patients and the formation of a cartilage-like ECM that can integrate into the surrounding tissue in vivo.


Subject(s)
Activins/metabolism , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Chondrocytes/pathology , Activins/genetics , Aged , Animals , Bone Morphogenetic Protein 2/genetics , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/transplantation , Collagen/genetics , Collagen/metabolism , Extracellular Matrix/metabolism , Female , Gene Expression , Humans , Immunohistochemistry , Ligands , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Osteoarthritis/pathology , Osteoarthritis/therapy , Proteoglycans/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transplantation, Autologous , Transplantation, Heterologous
6.
Stem Cells ; 32(2): 436-46, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24420904

ABSTRACT

Fanconi anemia (FA) is a complex genetic disease associated with a defective DNA repair pathway known as the FA pathway. In contrast to many other FA proteins, BRCA2 participates downstream in this pathway and has a critical role in homology-directed recombination (HDR). In our current studies, we have observed an extremely low reprogramming efficiency in cells with a hypomorphic mutation in Brca2 (Brca2(Δ) (27/) (Δ27)), that was associated with increased apoptosis and defective generation of nuclear RAD51 foci during the reprogramming process. Gene complementation facilitated the generation of Brca2(Δ) (27/) (Δ27) induced pluripotent stem cells (iPSCs) with a disease-free FA phenotype. Karyotype analyses and comparative genome hybridization arrays of complemented Brca2(Δ) (27/) (Δ27) iPSCs showed, however, the presence of different genetic alterations in these cells, most of which were not evident in their parental Brca2(Δ) (27/) (Δ27) mouse embryonic fibroblasts. Gene-corrected Brca2(Δ) (27/) (Δ27) iPSCs could be differentiated in vitro toward the hematopoietic lineage, although with a more limited efficacy than WT iPSCs or mouse embryonic stem cells, and did not engraft in irradiated Brca2(Δ) (27/) (Δ27) recipients. Our results are consistent with previous studies proposing that HDR is critical for cell reprogramming and demonstrate that reprogramming defects characteristic of Brca2 mutant cells can be efficiently overcome by gene complementation. Finally, based on analysis of the phenotype, genetic stability, and hematopoietic differentiation potential of gene-corrected Brca2(Δ) (27/) (Δ) (27) iPSCs, achievements and limitations in the application of current reprogramming approaches in hematopoietic stem cell therapy are also discussed.


Subject(s)
BRCA2 Protein/genetics , Fanconi Anemia/genetics , Genetic Therapy , Hematopoietic Stem Cells , Induced Pluripotent Stem Cells/cytology , Animals , BRCA2 Protein/biosynthesis , Cell Differentiation/genetics , Cells, Cultured , Cellular Reprogramming , DNA Damage/genetics , Fanconi Anemia/pathology , Fanconi Anemia/therapy , Fibroblasts/metabolism , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice
7.
Oncogene ; 31(39): 4333-42, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22179836

ABSTRACT

Angiogenesis is essential for development and tumor progression. With the aim of identifying new compound inhibitors of the angiogenesis process, we used an established enhanced green fluorescent protein-transgenic zebrafish line to develop an automated assay that enables high-throughput screening of compound libraries in a whole-organism setting. Using this system, we have identified novel kinase inhibitor compounds that show anti-angiogenic properties in both zebrafish in-vivo system and in human endothelial cell in-vitro angiogenesis models. Furthermore, we have determined the kinase target of these compounds and have identified and validated a previously uncharacterized involvement of phosphorylase kinase subunit G1 (PhKG1) in angiogenesis in vivo. In addition, we have found that PhKG1 is upregulated in human tumor samples and that aberrations in gene copy number of PhK subunits are a common feature of human tumors. Our results provide a novel insight into the angiogenesis process, as well as identify new potential targets for anti-angiogenic therapies.


Subject(s)
Angiogenesis Inhibitors/isolation & purification , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Phosphorylase Kinase/antagonists & inhibitors , Zebrafish , Angiogenesis Inhibitors/pharmacology , Animals , Animals, Genetically Modified , Cell Line , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Gene Dosage , High-Throughput Screening Assays , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Phosphorylase Kinase/genetics , Up-Regulation
8.
Cell Death Differ ; 19(1): 132-43, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21660050

ABSTRACT

The death inducer obliterator (Dido) locus encodes three protein isoforms, of which Dido3 is the largest and most broadly expressed. Dido3 is a nuclear protein that forms part of the spindle assembly checkpoint (SAC) and is necessary for correct chromosome segregation in somatic and germ cells. Here we report that specific ablation of Dido3 function in mice causes lethal developmental defects at the onset of gastrulation. Although these defects are associated with centrosome amplification, spindle malformation and a DNA damage response, we provide evidence that embryonic lethality of the Dido3 mutation cannot be explained by its impact on chromosome segregation alone. We show that loss of Dido3 expression compromises differentiation of embryonic stem cells in vitro and of epiblast cells in vivo, resulting in early embryonic death at around day 8.5 of gestation. Close analysis of Dido3 mutant embryoid bodies indicates that ablation of Dido3, rather than producing a generalized differentiation blockade, delays the onset of lineage commitment at the primitive endoderm specification stage. The dual role of Dido3 in chromosome segregation and stem cell differentiation supports the implication of SAC components in stem cell fate decisions.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Development/genetics , M Phase Cell Cycle Checkpoints/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Centrosome/metabolism , DNA Damage/genetics , Embryoid Bodies/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Endoderm/embryology , Gastrulation/genetics , Gene Expression Regulation, Developmental , Germ Layers/cytology , Germ Layers/growth & development , Mice , Mutation
9.
Hum Gene Ther ; 23(1): 56-69, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21877920

ABSTRACT

Human embryonic stem cells (hESC) and induced pluripotent stem cells (iPSC) offer great hope for in vitro modeling of Parkinson's disease (PD), as well as for designing cell-replacement therapies. To realize these opportunities, there is an urgent need to develop efficient protocols for the directed differentiation of hESC/iPSC into dopamine (DA) neurons with the specific characteristics of the cell population lost to PD, i.e., A9-subtype ventral midbrain DA neurons. Here we use lentiviral vectors to drive the expression of LMX1A, which encodes a transcription factor critical for ventral midbrain identity, specifically in neural progenitor cells. We show that clonal lines of hESC engineered to contain one or two copies of this lentiviral vector retain long-term self-renewing ability and pluripotent differentiation capacity. Greater than 60% of all neurons generated from LMX1A-engineered hESC were ventral midbrain DA neurons of the A9 subtype, compared with ∼10% in green fluorescent protein-engineered controls, as judged by specific marker expression and functional analyses. Moreover, DA neuron precursors differentiated from LMX1A-engineered hESC were able to survive and differentiate when grafted into the brain of adult mice. Finally, we provide evidence that LMX1A overexpression similarly increases the yield of DA neuron differentiation from human iPSC. Taken together, our data show that stable genetic engineering of hESC/iPSC with lentiviral vectors driving controlled expression of LMX1A is an efficient way to generate enriched populations of human A9-subtype ventral midbrain DA neurons, which should prove useful for modeling PD and may be helpful for designing future cell-replacement strategies.


Subject(s)
Dopaminergic Neurons/cytology , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , LIM-Homeodomain Proteins/metabolism , Lentivirus/metabolism , Transcription Factors/metabolism , Animals , Cell Count , Cell Differentiation , Cells, Cultured , Dopaminergic Neurons/metabolism , Embryonic Stem Cells/metabolism , Genetic Engineering/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/metabolism , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , LIM-Homeodomain Proteins/genetics , Lentivirus/genetics , Mesencephalon/cytology , Mesencephalon/metabolism , Mice , Mice, Nude , Mice, SCID , Plasmids/genetics , Plasmids/metabolism , Stem Cell Transplantation , Teratoma/pathology , Transcription Factors/genetics , Transgenes
10.
Stem Cells ; 29(8): 1186-95, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21681858

ABSTRACT

Gamete failure-derived infertility affects millions of people worldwide; for many patients, gamete donation by unrelated donors is the only available treatment. Embryonic stem cells (ESCs) can differentiate in vitro into germ-like cells, but they are genetically unrelated to the patient. Using an in vitro protocol that aims at recapitulating development, we have achieved, for the first time, complete differentiation of human induced pluripotent stem cells (hiPSCs) to postmeiotic cells. Unlike previous reports using human ESCs, postmeiotic cells arose without the over-expression of germline related transcription factors. Moreover, we consistently obtained haploid cells from hiPSCs of different origin (keratinocytes and cord blood), produced with a different number of transcription factors, and of both genetic sexes, suggesting the independence of our approach from the epigenetic memory of the reprogrammed somatic cells. Our work brings us closer to the production of personalized human gametes in vitro.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Meiosis , 3-Hydroxysteroid Dehydrogenases/metabolism , Adaptor Proteins, Signal Transducing , Antigens, CD/metabolism , Benzothiazoles/pharmacology , Cell Culture Techniques , Cell Cycle Proteins , Cell Differentiation/drug effects , Cell Line , Colforsin/pharmacology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA Methylation , DNA-Binding Proteins , Fibroblast Growth Factor 2/pharmacology , Gene Expression , Gene Expression Regulation , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Intermediate Filament Proteins/metabolism , Karyotyping , Leukemia Inhibitory Factor/pharmacology , Male , Nerve Tissue Proteins/metabolism , Nestin , Nuclear Proteins/metabolism , Ploidies , Promoter Regions, Genetic , Proteins/genetics , Proteins/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , Stage-Specific Embryonic Antigens/metabolism , Triazoles/pharmacology , Vimentin/metabolism
13.
Article in English | MEDLINE | ID: mdl-19028986

ABSTRACT

Human embryonic stem (hES) cells represent a potential source for cell replacement therapy of many degenerative diseases. Most frequently, hES cell lines are derived from surplus embryos from assisted reproduction cycles, independent of their quality or morphology. Here, we show that hES cell lines can be obtained from poor-quality blastocysts with the same efficiency as that obtained from good- or intermediate-quality blastocysts. Furthermore, we show that the self-renewal, pluripotency, and differentiation ability of hES cell lines derived from either source are comparable. Finally, we present a simple and reproducible embryoid body-based protocol for the differentiation of hES cells into functional cardiomyocytes. The five new hES cell lines derived here should widen the spectrum of available resources for investigating the biology of hES cells and advancing toward efficient strategies of regenerative medicine.


Subject(s)
Blastocyst/cytology , Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Biomarkers/metabolism , Cell Culture Techniques/methods , Cell Differentiation , Cell Line , Cell Proliferation , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Histocompatibility Testing , Humans , Karyotyping , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/immunology , Pluripotent Stem Cells/metabolism
14.
Annu Rev Cell Dev Biol ; 17: 87-132, 2001.
Article in English | MEDLINE | ID: mdl-11687485

ABSTRACT

Vertebrate limb buds are embryonic structures for which much molecular and cellular data are known regarding the mechanisms that control pattern formation during development. Specialized regions of the developing limb bud, such as the zone of polarizing activity (ZPA), the apical ectodermal ridge (AER), and the non-ridge ectoderm, direct and coordinate the development of the limb bud along the anterior-posterior (AP), dorsal-ventral (DV), and proximal-distal (PD) axes, giving rise to a stereotyped pattern of elements well conserved among tetrapods. In recent years, specific gene functions have been shown to mediate the organizing and patterning activities of the ZPA, the AER, and the non-ridge ectoderm. The analysis of these gene functions has revealed the existence of complex interactions between signaling pathways operated by secreted factors of the HH, TGF-beta/BMP, WNT, and FGF superfamilies, which interact with many other genetic networks to control limb positioning, outgrowth, and patterning. The study of limb development has helped to establish paradigms for the analysis of pattern formation in many other embryonic structures and organs.


Subject(s)
Body Patterning/genetics , Extremities/embryology , Trans-Activators/genetics , Animals , Cell Differentiation/genetics , Ectoderm , Embryonic Induction , Epithelium/embryology , Gene Expression Regulation, Developmental , Hedgehog Proteins , Organizers, Embryonic , Signal Transduction
15.
Dev Cell ; 1(3): 423-34, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11702953

ABSTRACT

Dickkopf1 (Dkk1) is a secreted protein that acts as a Wnt inhibitor and, together with BMP inhibitors, is able to induce the formation of ectopic heads in Xenopus. Here, we show that Dkk1 null mutant embryos lack head structures anterior of the midbrain. Analysis of chimeric embryos implicates the requirement of Dkk1 in anterior axial mesendoderm but not in anterior visceral endoderm for head induction. In addition, mutant embryos show duplications and fusions of limb digits. Characterization of the limb phenotype strongly suggests a role for Dkk1 both in cell proliferation and in programmed cell death. Our data provide direct genetic evidence for the requirement of secreted Wnt antagonists during embryonic patterning and implicate Dkk1 as an essential inducer during anterior specification as well as a regulator during distal limb patterning.


Subject(s)
Embryo, Mammalian/physiology , Embryonic Induction/physiology , Extremities/embryology , Head/embryology , Morphogenesis/physiology , Proteins/metabolism , Zebrafish Proteins , Animals , Biomarkers , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Brain/embryology , Chick Embryo , Embryo, Mammalian/ultrastructure , Extremities/growth & development , Gene Targeting , Head/growth & development , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Molecular Sequence Data , Proteins/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Wnt Proteins
16.
Development ; 128(16): 3189-95, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11688567

ABSTRACT

Expression of the Nodal gene, which encodes a member of the TGFbeta superfamily of secreted factors, localizes to the left side of the developing embryo in all vertebrates examined so far. This asymmetric pattern correlates with normal development of the left-right axis. We now show that the Wnt and PKA signaling pathways control left-right determination in the chick embryo through Nodal. A Wnt/beta-catenin pathway controls Nodal expression in and around Hensen's node, without affecting the upstream regulators Sonic hedgehog, Car and Fibroblast Growth Factor 8. Transcription of Nodal is also positively regulated by a protein kinase A-dependent pathway. Both the adhesion protein N-cadherin and PKI (an endogenous protein kinase A inhibitor) are localized to the right side of the node and may contribute to restrict Nodal activation by Wnt signaling and PKA to the left side of the node.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Proto-Oncogene Proteins/physiology , Transforming Growth Factor beta/biosynthesis , Zebrafish Proteins , Alkaline Phosphatase/metabolism , Animals , Cadherins/biosynthesis , Cell Adhesion Molecules/biosynthesis , Chick Embryo , Embryonic Induction , Gene Expression Regulation, Developmental , Hedgehog Proteins , In Situ Hybridization , Models, Biological , Nodal Protein , Signal Transduction , Trans-Activators/biosynthesis , Transforming Growth Factor beta/metabolism , Wnt Proteins
17.
Cell ; 104(6): 891-900, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11290326

ABSTRACT

A regulatory loop between the fibroblast growth factors FGF-8 and FGF-10 plays a key role in limb initiation and AER induction in vertebrate embryos. Here, we show that three WNT factors signaling through beta-catenin act as key regulators of the FGF-8/FGF-10 loop. The Wnt-2b gene is expressed in the intermediate mesoderm and the lateral plate mesoderm in the presumptive chick forelimb region. Cells expressing Wnt-2b are able to induce Fgf-10 and generate an extra limb when implanted into the flank. In the presumptive hindlimb region, another Wnt gene, Wnt-8c, controls Fgf-10 expression, and is also capable of inducing ectopic limb formation in the flank. Finally, we also show that the induction of Fgf-8 in the limb ectoderm by FGF-10 is mediated by the induction of Wnt-3a. Thus, three WNT signals mediated by beta-catenin control both limb initiation and AER induction in the vertebrate embryo.


Subject(s)
Chick Embryo/physiology , Fibroblast Growth Factors/physiology , Limb Buds/physiology , Proto-Oncogene Proteins/physiology , Trans-Activators , Zebrafish Proteins , Animals , Avian Proteins , Cytoskeletal Proteins/physiology , Ectoderm/physiology , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Mesoderm/physiology , Mitogens/physiology , Morphogenesis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Signal Transduction , Wings, Animal/embryology , Wnt Proteins , Wnt2 Protein , beta Catenin
18.
Curr Opin Cell Biol ; 13(2): 204-10, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11248554

ABSTRACT

Appendages, such as wings of a fly or limbs of a vertebrate, are excellent models to study the principles of patterning and morphogenesis. In the adult these structures are used for a variety of behaviors, including locomotion. Although support structures of the adult vertebrate limb are generated within the limb bud, its dynamic elements are derived from the somitic mesoderm and neural tube. Recent studies show that regional patterns set up in the mesenchyme-filled limb bud guide muscle precursors and developing motor axons to their proper location within the limb. Subsequent development of the neuromuscular system is regulated by cell surface interactions between pre-specified muscle fibers and motor axons.


Subject(s)
Extremities/embryology , Muscles/embryology , Nervous System/embryology , Animals , Extremities/innervation , Humans , Motor Neurons/physiology , Muscles/physiology
19.
IUBMB Life ; 50(1): 1-11, 2000 Jul.
Article in English | MEDLINE | ID: mdl-11087114

ABSTRACT

Vertebrates exhibit evolutionarily conserved asymmetries in the pattern of internal organ placement that are essential for their normal physiological function. Left-right asymmetries in organ situs are dependent upon the formation of an intact left-right axis during embryogenesis. Recently many of the molecular components involved in the initiation and maintenance of the left-right axis have been described. These molecules and their function in promoting left-right asymmetries are reviewed.


Subject(s)
Body Patterning/physiology , Embryo, Mammalian/physiology , Embryo, Nonmammalian , Vertebrates/embryology , Animals , Biological Evolution , Chick Embryo , Functional Laterality , Gene Expression Regulation, Developmental , Mice , Signal Transduction , Transcription Factors/physiology , Transforming Growth Factor beta/metabolism
20.
Development ; 127(23): 5101-12, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11060236

ABSTRACT

The vertebrate brain develops from a bilaterally symmetric neural tube but later displays profound anatomical and functional asymmetries. Despite considerable progress in deciphering mechanisms of visceral organ laterality, the genetic pathways regulating brain asymmetries are unknown. In zebrafish, genes implicated in laterality of the viscera (cyclops/nodal, antivin/lefty and pitx2) are coexpressed on the left side of the embryonic dorsal diencephalon, within a region corresponding to the presumptive epiphysis or pineal organ. Asymmetric gene expression in the brain requires an intact midline and Nodal-related factors. RNA-mediated rescue of mutants defective in Nodal signaling corrects tissue patterning at gastrulation, but fails to restore left-sided gene expression in the diencephalon. Such embryos develop into viable adults with seemingly normal brain morphology. However, the pineal organ, which typically emanates at a left-to-medial site from the dorsal diencephalic roof, becomes displaced in position. Thus, a conserved signaling pathway regulating visceral laterality also underlies an anatomical asymmetry of the zebrafish forebrain.


Subject(s)
Body Patterning/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Nuclear Proteins , Pineal Gland/embryology , Signal Transduction/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Zebrafish Proteins , Animals , Brain/embryology , Diencephalon/embryology , Endoderm , Epiphyses , Female , Intracellular Signaling Peptides and Proteins , Left-Right Determination Factors , Male , Mutagenesis , Nodal Protein , Paired Box Transcription Factors , Prosencephalon/embryology , Zebrafish/embryology , Zebrafish/genetics , Homeobox Protein PITX2
SELECTION OF CITATIONS
SEARCH DETAIL
...