Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Commun Biol ; 6(1): 611, 2023 06 07.
Article in English | MEDLINE | ID: mdl-37286713

ABSTRACT

Although neural stem/progenitor cells derived from human induced pluripotent stem cells (hiPSC-NS/PCs) are expected to be a cell source for cell-based therapy, tumorigenesis of hiPSC-NS/PCs is a potential problem for clinical applications. Therefore, to understand the mechanisms of tumorigenicity in NS/PCs, we clarified the cell populations of NS/PCs. We established single cell-derived NS/PC clones (scNS/PCs) from hiPSC-NS/PCs that generated undesired grafts. Additionally, we performed bioassays on scNS/PCs, which classified cell types within parental hiPSC-NS/PCs. Interestingly, we found unique subsets of scNS/PCs, which exhibited the transcriptome signature of mesenchymal lineages. Furthermore, these scNS/PCs expressed both neural (PSA-NCAM) and mesenchymal (CD73 and CD105) markers, and had an osteogenic differentiation capacity. Notably, eliminating CD73+ CD105+ cells from among parental hiPSC-NS/PCs ensured the quality of hiPSC-NS/PCs. Taken together, the existence of unexpected cell populations among NS/PCs may explain their tumorigenicity leading to potential safety issues of hiPSC-NS/PCs for future regenerative medicine.


Subject(s)
Induced Pluripotent Stem Cells , Neural Stem Cells , Humans , Osteogenesis , Neural Stem Cells/metabolism , Cell Transformation, Neoplastic/metabolism , Carcinogenesis/metabolism
2.
Stem Cells Transl Med ; 10(3): 398-413, 2021 03.
Article in English | MEDLINE | ID: mdl-33226180

ABSTRACT

Cell-based therapy targeting spinal cord injury (SCI) is an attractive approach to promote functional recovery by replacing damaged tissue. We and other groups have reported the effectiveness of transplanting neural stem/progenitor cells (NS/PCs) derived from human induced pluripotent stem cells (hiPSCs) in SCI animal models for neuronal replacement. Glial replacement is an additional approach for tissue repair; however, the lack of robust procedures to drive iPSCs into NS/PCs which can produce glial cells has hindered the development of glial cell transplantation for the restoration of neuronal functions after SCI. Here, we established a method to generate NS/PCs with gliogenic competence (gNS/PCs) optimized for clinical relevance and utilized them as a source of therapeutic NS/PCs for SCI. We could successfully generate gNS/PCs from clinically relevant hiPSCs, which efficiently produced astrocytes and oligodendrocytes in vitro. We also performed comparison between gNS/PCs and neurogenic NS/PCs based on single cell RNA-seq analysis and found that gNS/PCs were distinguished by expression of several transcription factors including HEY2 and NFIB. After gNS/PC transplantation, the graft did not exhibit tumor-like tissue formation, indicating the safety of them as a source of cell therapy. Importantly, the gNS/PCs triggered functional recovery in an SCI animal model, with remyelination of demyelinated axons and improved motor function. Given the inherent safety of gNS/PCs and favorable outcomes observed after their transplantation, cell-based medicine using the gNS/PCs-induction procedure described here together with clinically relevant iPSCs is realistic and would be beneficial for SCI patients.


Subject(s)
Cell Culture Techniques , Induced Pluripotent Stem Cells , Neural Stem Cells , Spinal Cord Injuries , Animals , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/transplantation , Neural Stem Cells/transplantation , Recovery of Function , Spinal Cord , Spinal Cord Injuries/therapy , Stem Cell Transplantation
3.
Antioxidants (Basel) ; 9(5)2020 May 14.
Article in English | MEDLINE | ID: mdl-32422904

ABSTRACT

Amyotrophic lateral sclerosis and Parkinsonism-dementia complex (ALS/PDC) is a unique endemic neurodegenerative disease, with high-incidence foci in Kii Peninsula, Japan. To gather new insights into the pathological mechanisms underlying Kii ALS/PDC, we performed transcriptome analyses of patient brains. We prepared frozen brains from three individuals without neurodegenerative diseases, three patients with Alzheimer's disease, and 21 patients with Kii ALS/PDC, and then acquired microarray data from cerebral gray and white matter tissues. Microarray results revealed that expression levels of genes associated with heat shock proteins, DNA binding/damage, and senescence were significantly altered in patients with ALS/PDC compared with healthy individuals. The RNA expression pattern observed for ALS-type brains was similar to that of PDC-type brains. Additionally, pathway and network analyses indicated that the molecular mechanism underlying ALS/PDC may be associated with oxidative phosphorylation of mitochondria, ribosomes, and the synaptic vesicle cycle; in particular, upstream regulators of these mechanisms may be found in synapses and during synaptic trafficking. Furthermore, phenotypic differences between ALS-type and PDC-type were observed, based on HLA haplotypes. In conclusion, determining the relationship between stress-responsive proteins, synaptic dysfunction, and the pathogenesis of ALS/PDC in the Kii peninsula may provide new understanding of this mysterious disease.

4.
Stem Cells ; 37(1): 6-13, 2019 01.
Article in English | MEDLINE | ID: mdl-30371964

ABSTRACT

There have been numerous attempts to develop stem cell transplantation approaches to promote the regeneration of spinal cord injury (SCI). Our multicenter team is currently planning to launch a first-in-human clinical study of an induced pluripotent stem cell (iPSC)-based cell transplant intervention for subacute SCI. This trial was conducted as class I regenerative medicine protocol as provided for under Japan's Act on the Safety of Regenerative Medicine, using neural stem/progenitor cells derived from a clinical-grade, integration-free human "iPSC stock" generated by the Kyoto University Center for iPS Cell Research and Application. In the present article, we describe how we are preparing to initiate this clinical study, including addressing the issues of safety and tumorigenesis as well as practical problems that must be overcome to enable the development of therapeutic interventions for patients with chronic SCI. Stem Cells 2019;37:6-13.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Spinal Cord Injuries/therapy , Stem Cell Transplantation/methods , Humans
5.
Stem Cell Res ; 19: 128-138, 2017 03.
Article in English | MEDLINE | ID: mdl-28135684

ABSTRACT

To achieve the goal of a first-in-human trial for human induced pluripotent stem cell (hiPSC)-based transplantation for the treatment of various diseases, allogeneic human leukocyte antigen (HLA)-matched hiPSC cell banks represent a realistic tool from the perspective of quality control and cost performance. Furthermore, considering the limited therapeutic time-window for acute injuries, including neurotraumatic injuries, an iPS cell bank is of potential interest. However, due to the relatively immunoprivileged environment of the central nervous system, it is unclear whether HLA matching is required in hiPSC-derived neural stem/progenitor cell (hiPSC-NS/PC) transplantation for the treatment of neurodegenerative diseases and neurotraumatic injuries. In this study, we evaluated the significance of HLA matching in hiPSC-NS/PC transplantation by performing modified mixed lymphocyte reaction (MLR) assays with hiPSC-NS/PCs. Compared to fetus-derived NS/PCs, the expression levels of human leukocyte antigen-antigen D related (HLA-DR) and co-stimulatory molecules on hiPSC-NS/PCs were significantly low, even with the addition of tumor necrosis factor-α (TNFα) and/or interferon-γ (IFNγ) to mimic the inflammatory environment surrounding transplanted hiPSC-NS/PCs in injured tissues. Interestingly, both the allogeneic HLA-matched and the HLA-mismatched responses were similarly low in the modified MLR assay. Furthermore, the autologous response was also similar to the allogeneic response. hiPSC-NS/PCs suppressed the proliferative responses of allogeneic HLA-mismatched peripheral blood mononuclear cells (PBMCs) in a dose-dependent manner. Thus, the low antigen-presenting function and immunosuppressive effects of hiPSC-NS/PCs result in a depressed immune response, even in an allogeneic HLA-mismatched setting. It is crucial to verify whether these in vitro results are reproducible in a clinical setting.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/immunology , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cells, Cultured , Coculture Techniques , Fetus/cytology , Gene Expression/drug effects , Genotype , HLA-DR Antigens/genetics , HLA-DR Antigens/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Interferon-gamma/pharmacology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Tumor Necrosis Factor-alpha/pharmacology
6.
Mol Brain ; 9(1): 88, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27716287

ABSTRACT

Patient-specific induced pluripotent stem cells (iPSCs) facilitate understanding of the etiology of diseases, discovery of new drugs and development of novel therapeutic interventions. A frequently used starting source of cells for generating iPSCs has been dermal fibroblasts (DFs) isolated from skin biopsies. However, there are also numerous repositories containing lymphoblastoid B-cell lines (LCLs) generated from a variety of patients. To date, this rich bioresource of LCLs has been underused for generating iPSCs, and its use would greatly expand the range of targeted diseases that could be studied by using patient-specific iPSCs. However, it remains unclear whether patient's LCL-derived iPSCs (LiPSCs) can function as a disease model. Therefore, we generated Parkinson's disease patient-specific LiPSCs and evaluated their utility as tools for modeling neurological diseases. We established iPSCs from two LCL clones, which were derived from a healthy donor and a patient carrying PARK2 mutations, by using existing non-integrating episomal protocols. Whole genome sequencing (WGS) and comparative genomic hybridization (CGH) analyses showed that the appearance of somatic variations in the genomes of the iPSCs did not vary substantially according to the original cell types (LCLs, T-cells and fibroblasts). Furthermore, LiPSCs could be differentiated into functional neurons by using the direct neurosphere conversion method (dNS method), and they showed several Parkinson's disease phenotypes that were similar to those of DF-iPSCs. These data indicate that the global LCL repositories can be used as a resource for generating iPSCs and disease models. Thus, LCLs are the powerful tools for generating iPSCs and modeling neurological diseases.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Lymphocytes/cytology , Models, Biological , Nervous System Diseases/pathology , Base Sequence , Cell Differentiation , Cell Line, Transformed , Dermis/cytology , Fibroblasts/cytology , Herpesvirus 4, Human/physiology , Humans , Mitochondria/metabolism , Mutation/genetics , Neurons/cytology , Parkinson Disease/pathology , Phenotype , Spheroids, Cellular/cytology , Ubiquitin-Protein Ligases/metabolism
7.
Mol Brain ; 9(1): 85, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27642008

ABSTRACT

The risk of tumorigenicity is a hurdle for regenerative medicine using induced pluripotent stem cells (iPSCs). Although teratoma formation is readily distinguishable, the malignant transformation of iPSC derivatives has not been clearly defined due to insufficient analysis of histology and phenotype. In the present study, we evaluated the histology of neural stem/progenitor cells (NSPCs) generated from integration-free human peripheral blood mononuclear cell (PBMC)-derived iPSCs (iPSC-NSPCs) following transplantation into central nervous system (CNS) of immunodeficient mice. We found that transplanted iPSC-NSPCs produced differentiation patterns resembling those in embryonic CNS development, and that the microenvironment of the final site of migration affected their maturational stage. Genomic instability of iPSCs correlated with increased proliferation of transplants, although no carcinogenesis was evident. The histological classifications presented here may provide cues for addressing potential safety issues confronting regenerative medicine involving iPSCs.


Subject(s)
Central Nervous System Diseases/therapy , Induced Pluripotent Stem Cells/pathology , Neural Stem Cells/pathology , Stem Cell Transplantation/adverse effects , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Central Nervous System Diseases/pathology , Genomic Instability , Humans , Induced Pluripotent Stem Cells/transplantation , Karyotype , Mice, SCID , Models, Biological , Neural Stem Cells/transplantation , Registries
8.
Neurosci Res ; 110: 18-28, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27083781

ABSTRACT

Neural stem/progenitor cells (NS/PCs) derived from human induced pluripotent stem cells (hiPSCs) are expected to be a valuable cell source for cell therapies that target central nervous system disorders. For clinical applications, NS/PCs should be induced and maintained under clinical grade conditions, which are challenging to achieve. In the present study, we established a procedure to obtain xeno-free long-term self-renewing neuroepithelial-like stem cells (xf-lt-NES cells) from feeder-free hiPSCs using a newly developed xeno-free medium, StemFit(®)AS200. xf-lt-NES cells were cultured for long periods in StemFit(®)AS200 while retaining normal karyotypes, NS/PC marker expression and differentiation capacity for neuronal and glial differentiation in vitro and in vivo. Furthermore, the cells were cryopreserved using a defined serum-free freezing reagent, which demonstrated the feasibility of this xeno-free culture system for large-scale lt-NES cell production and cell banking. Taken together, our system represents a promising approach for the manufacture of clinically relevant products for cell therapy using NS/PCs.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Leukocytes, Mononuclear/cytology , Neural Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Lineage , Culture Media , Female , Humans , Mice , Neural Stem Cells/transplantation , Neuroglia/cytology , Neurons/cytology
9.
Stem Cell Reports ; 6(3): 422-35, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26905201

ABSTRACT

Modeling of neurological diseases using induced pluripotent stem cells (iPSCs) derived from the somatic cells of patients has provided a means of elucidating pathogenic mechanisms and performing drug screening. T cells are an ideal source of patient-specific iPSCs because they can be easily obtained from samples. Recent studies indicated that iPSCs retain an epigenetic memory relating to their cell of origin that restricts their differentiation potential. The classical method of differentiation via embryoid body formation was not suitable for T cell-derived iPSCs (TiPSCs). We developed a neurosphere-based robust differentiation protocol, which enabled TiPSCs to differentiate into functional neurons, despite differences in global gene expression between TiPSCs and adult human dermal fibroblast-derived iPSCs. Furthermore, neurons derived from TiPSCs generated from a juvenile patient with Parkinson's disease exhibited several Parkinson's disease phenotypes. Therefore, we conclude that TiPSCs are a useful tool for modeling neurological diseases.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neurogenesis , Neurons/cytology , Primary Cell Culture/methods , T-Lymphocytes/cytology , Cells, Cultured , Fibroblasts/cytology , Humans
10.
Stem Cell Reports ; 6(1): 1-8, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26724902

ABSTRACT

Murine- and human-induced pluripotent stem cell-derived neural stem/progenitor cells (iPSC-NS/PCs) promote functional recovery following transplantation into the injured spinal cord in rodents and primates. Although remyelination of spared demyelinated axons is a critical mechanism in the regeneration of the injured spinal cord, human iPSC-NS/PCs predominantly differentiate into neurons both in vitro and in vivo. We therefore took advantage of our recently developed protocol to obtain human-induced pluripotent stem cell-derived oligodendrocyte precursor cell-enriched neural stem/progenitor cells and report the benefits of transplanting these cells in a spinal cord injury (SCI) model. We describe how this approach contributes to the robust remyelination of demyelinated axons and facilitates functional recovery after SCI.


Subject(s)
Axons/physiology , Induced Pluripotent Stem Cells/transplantation , Myelin Sheath/physiology , Oligodendroglia/physiology , Spinal Cord Injuries/therapy , Animals , Cell Differentiation/physiology , Cells, Cultured , Female , Hindlimb/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Mice, Inbred NOD , Mice, SCID , Nerve Regeneration/physiology , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Neurons/cytology , Neurons/physiology , Oligodendroglia/cytology , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Stem Cell Transplantation/methods , Transplantation, Heterologous
11.
Neurosci Res ; 107: 20-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26804710

ABSTRACT

Stem cells represent a potential cellular resource in the development of regenerative medicine approaches to the treatment of pathologies in which specific cells are degenerated or damaged by genetic abnormality, disease, or injury. Securing sufficient supplies of cells suited to the demands of cell transplantation, however, remains challenging, and the establishment of safe and efficient cell banking procedures is an important goal. Cryopreservation allows the storage of stem cells for prolonged time periods while maintaining them in adequate condition for use in clinical settings. Conventional cryopreservation systems include slow-freezing and vitrification both have advantages and disadvantages in terms of cell viability and/or scalability. In the present study, we developed an advanced slow-freezing technique using a programmed freezer with a magnetic field called Cells Alive System (CAS) and examined its effectiveness on human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NS/PCs). This system significantly increased cell viability after thawing and had less impact on cellular proliferation and differentiation. We further found that frozen-thawed hiPSC-NS/PCs were comparable with non-frozen ones at the transcriptome level. Given these findings, we suggest that the CAS is useful for hiPSC-NS/PCs banking for clinical uses involving neural disorders and may open new avenues for future regenerative medicine.


Subject(s)
Cryopreservation/instrumentation , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Cell Survival , Cells, Cultured , Cryopreservation/methods , Humans , Magnetic Fields
12.
Dev Biol ; 391(2): 196-206, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24768892

ABSTRACT

The broad diversity of neurons is vital to neuronal functions. During vertebrate development, the spinal cord is a site of sensory and motor tasks coordinated by interneurons and the ongoing neurogenesis. In the spinal cord, V2-interneuron (V2-IN) progenitors (p2) develop into excitatory V2a-INs and inhibitory V2b-INs. The balance of these two types of interneurons requires precise control in the number and timing of their production. Here, using zebrafish embryos with altered Notch signaling, we show that different combinations of Notch ligands and receptors regulate two functions: the maintenance of p2 progenitor cells and the V2a/V2b cell fate decision in V2-IN development. Two ligands, DeltaA and DeltaD, and three receptors, Notch1a, Notch1b, and Notch3 redundantly contribute to p2 progenitor maintenance. On the other hand, DeltaA, DeltaC, and Notch1a mainly contribute to the V2a/V2b cell fate determination. A ubiquitin ligase Mib, which activates Notch ligands, acts in both functions through its activation of DeltaA, DeltaC, and DeltaD. Moreover, p2 progenitor maintenance and V2a/V2b fate determination are not distinct temporal processes, but occur within the same time frame during development. In conclusion, V2-IN cell progenitor proliferation and V2a/V2b cell fate determination involve signaling through different sets of Notch ligand-receptor combinations that occur concurrently during development in zebrafish.


Subject(s)
Interneurons/cytology , Neural Stem Cells/cytology , Neurogenesis/genetics , Receptors, Notch/genetics , Spinal Cord/embryology , Zebrafish/embryology , Animals , Cell Proliferation , Gene Expression Regulation, Developmental , Gene Knockout Techniques/veterinary , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Morpholinos/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Receptor, Notch1/metabolism , Receptor, Notch3 , Receptors, Notch/metabolism , Signal Transduction/genetics , Spinal Cord/cytology , Ubiquitin-Protein Ligases/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
Development ; 137(15): 2527-37, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20573700

ABSTRACT

In the developing embryo, cell-cell signalling is necessary for tissue patterning and structural organization. During midline development, the notochord plays roles in the patterning of its surrounding tissues while forming the axial structure; however, how these patterning and structural roles are coordinated remains elusive. Here, we identify a mechanism by which Notch signalling regulates the patterning activities and structural integrity of the notochord. We found that Mind bomb (Mib) ubiquitylates Jagged 1 (Jag1) and is essential in the signal-emitting cells for Jag1 to activate Notch signalling. In zebrafish, loss- and gain-of-function analyses showed that Mib-Jag1-Notch signalling favours the development of non-vacuolated cells at the expense of vacuolated cells in the notochord. This leads to changes in the peri-notochordal basement membrane formation and patterning surrounding the muscle pioneer cells. These data reveal a previously unrecognized mechanism regulating the patterning and structural roles of the notochord by Mib-Jag1-Notch signalling-mediated cell-fate determination.


Subject(s)
Body Patterning , Calcium-Binding Proteins/metabolism , Cell Lineage , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Notochord/physiology , Receptors, Notch/metabolism , Ubiquitin-Protein Ligases/metabolism , Zebrafish Proteins/metabolism , 3T3 Cells , Animals , COS Cells , Chlorocebus aethiops , Endocytosis , Jagged-1 Protein , Mice , Models, Biological , Serrate-Jagged Proteins , Two-Hybrid System Techniques , Ubiquitin/metabolism , Zebrafish
14.
Nat Cell Biol ; 12(3): 278-85, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20118921

ABSTRACT

The Notch signalling pathway has a crucial function in determining cell fates in multiple tissues within metazoan organisms. On binding to ligands, the Notch receptor is cleaved proteolytically and releases its intracellular domain (NotchICD). The NotchICD enters the nucleus and acts cooperatively with other factors to stimulate the transcription of target genes. High levels of Notch-mediated transcriptional activation require the formation of a ternary complex consisting of NotchICD, CSL (CBF-1, suppressor of hairless, LAG-1) and a Mastermind family member. However, it is still not clear how the formation of the ternary complex is regulated. Here we show that Nemo-like kinase (NLK) negatively regulates Notch-dependent transcriptional activation by decreasing the formation of this ternary complex. Using a biochemical screen, we identified Notch as a new substrate of NLK. NLK-phosphorylated Notch1ICD is impaired in its ability to form a transcriptionally active ternary complex. Furthermore, knockdown of NLK leads to hyperactivation of Notch signalling and consequently decreases neurogenesis in zebrafish. Our results both define a new function for NLK and reveal a previously unidentified mode of regulation in the Notch signalling pathway.


Subject(s)
Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Amino Acid Substitution/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , DNA/metabolism , ELAV Proteins/metabolism , ELAV-Like Protein 3 , Embryo, Nonmammalian/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mitogen-Activated Protein Kinases/genetics , Models, Biological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotides, Antisense/genetics , Phosphorylation/physiology , Protein Binding/physiology , Protein Interaction Domains and Motifs/physiology , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Notch/genetics , Transcription Factor HES-1 , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Xenopus , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...