Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Publication year range
1.
Med Mol Morphol ; 55(3): 199-209, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35578118

ABSTRACT

We investigated whether BMP4, FGF8, and/or WNT3a on neural crest-like cells (NCLC) derived from mouse induced pluripotent stem (miPS) cells will promote differentiation of odontoblasts-like cells. After the miPS cells matured into embryonic body (EB) cells, they were cultured in a neural induction medium to produce NCLC. As the differentiation of NCLC were confirmed by RT-qPCR, they were then disassociated and cultured with a medium containing, BMP4, FGF8, and/or WNT3a for 7 and 14 days. The effect of these stimuli on NCLC were assessed by RT-qPCR, ALP staining, and immunocytochemistry. The cultured EB cells presented a significant increase of Snai1, Slug, and Sox 10 substantiating the differentiation of NCLC. NCLC stimulated with more than two stimuli significantly increased the odontoblast markers Dmp-1, Dspp, Nestin, Alp, and Runx2 expression compared to control with no stimulus. The expression of Dmp-1 and Dspp upregulated more when FGF8 was combined with WNT3a. ALP staining was positive in groups containing BMP4 and fluorescence was observed in immunocytochemistry of the common significant groups between Dmp-1 and Dspp. After stimulation, the cell morphology demonstrated a spindle-shaped cells with long projections resembling odontoblasts. Simultaneous BMP4, FGF8, and WNT3a stimuli significantly differentiated NCLC into odontoblast-like cells.


Subject(s)
Bone Morphogenetic Protein 4 , Fibroblast Growth Factor 8 , Induced Pluripotent Stem Cells , Odontoblasts , Animals , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation , Cells, Cultured , Fibroblast Growth Factor 8/pharmacology , Induced Pluripotent Stem Cells/physiology , Mice , Neural Crest , Odontoblasts/metabolism , Wnt3A Protein/pharmacology
2.
Exp Eye Res ; 173: 129-137, 2018 08.
Article in English | MEDLINE | ID: mdl-29753729

ABSTRACT

Emerging therapies have begun to evaluate the abilities of Müller glial cells (MGCs) to protect and/or regenerate neurons following retina injury. The migration of donor cells is central to many reparative strategies, where cells must achieve appropriate positioning to facilitate localized repair. Although chemical cues have been implicated in the MGC migratory responses of numerous retinopathies, MGC-based therapies have yet to explore the extent to which external biochemical stimuli can direct MGC behavior. The current study uses a microfluidics-based assay to evaluate the migration of cultured rMC-1 cells (as model MGC) in response to quantitatively-controlled microenvironments of signaling factors implicated in retinal regeneration: basic Fibroblast Growth factor (bFGF or FGF2); Fibroblast Growth factor 8 (FGF8); Vascular Endothelial Growth Factor (VEGF); and Epidermal Growth Factor (EGF). Findings indicate that rMC-1 cells exhibited minimal motility in response to FGF2, FGF8 and VEGF, but highly-directional migration in response to EGF. Further, the responses were blocked by inhibitors of EGF-R and of the MAPK signaling pathway. Significantly, microfluidics data demonstrate that changes in the EGF gradient (i.e. change in EGF concentration over distance) resulted in the directional chemotactic migration of the cells. By contrast, small increases in EGF concentration, alone, resulted in non-directional cell motility, or chemokinesis. This microfluidics-enhanced approach, incorporating the ability both to modulate and asses the responses of motile donor cells to a range of potential chemotactic stimuli, can be applied to potential donor cell populations obtained directly from human specimens, and readily expanded to incorporate drug-eluting biomaterials and combinations of desired ligands.


Subject(s)
Chemotaxis/physiology , Ependymoglial Cells/physiology , Animals , Cell Proliferation/physiology , Cells, Cultured , Cellular Microenvironment , Ependymoglial Cells/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 8/pharmacology , Glial Fibrillary Acidic Protein/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Microfluidic Analytical Techniques , Nestin/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Receptors, Growth Factor/genetics , Vascular Endothelial Growth Factor A/pharmacology
3.
Tumour Biol ; 39(3): 1010428317695969, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28345462

ABSTRACT

Fibroblast growth factor 8b and androgen play important roles in cell proliferation of prostate cancer. We investigated the effects of fibroblast growth factor 8b and androgen on the proliferation of prostate cell lines and the corresponding intracellular mechanisms. It is found that dihydrotestosterone and fibroblast growth factor 8b stimulated Lncap cell mitosis in a concentration-responsive manner, with 30 ng/mL as the most suitable concentration, respectively. Dihydrotestosterone treatment alone did not enhance the expression and phosphorylation level of fibroblast growth factor receptor but significantly enhanced the level of fibroblast growth factor receptor phosphorylation elicited by fibroblast growth factor 8b. Phosphorylations of extracellular signal-regulated kinase, p38, and c-Jun NH2-terminal kinase were stimulated by dihydrotestosterone or fibroblast growth factor 8b. Among these major downstream pathways for mitogen-activated protein kinase, c-Jun NH2-terminal kinase signaling was most significantly enhanced. Protein kinase C phosphorylation was higher than AKT by the combined stimulation of dihydrotestosterone and fibroblast growth factor 8b. The phosphorylation of CDC2 was significantly induced by dihydrotestosterone and fibroblast growth factor 8b synergetically, and Smad underwent the same induction as CDC2. So the promoting effect of fibroblast growth factor 8b on cell cycle might contribute to the G2/M transition. This study indicated that the functional interaction between fibroblast growth factor 8b and androgen was essential for the prostate cancer cell proliferation.


Subject(s)
Cell Proliferation/drug effects , Dihydrotestosterone/pharmacology , Fibroblast Growth Factor 8/pharmacology , G2 Phase/drug effects , Prostatic Neoplasms/pathology , CDC2 Protein Kinase , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 8/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Phosphorylation/drug effects , Smad Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Mol Cell Biochem ; 425(1-2): 77-84, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27804049

ABSTRACT

Fibroblast growth factors (FGFs) comprise a large family of signaling molecules that involve cell patterning, mobilization, differentiation, and proliferation. Various FGFs, including FGF-1, FGF-2, and FGF-5, have been shown to play a role in cytoprotection during adverse cardiac events; however, whether FGF-8 is a cytoprotective remains unclear. The current study was designed to evaluate the effect of FGF-8 treatment on oxidative stress-induced apoptosis in H9c2 cells. Cells were divided into three groups: control, H2O2 (400 µm H2O2), and H2O2 + FGF-8 (4 ng/ml FGF-8). Our results suggest apoptosis was significantly (p < 0.05) enhanced in the H2O2 group relative to control. Moreover, a significant (p < 0.05) decline in apoptosis was observed in the H2O2 + FGF-8 group compared to H2O2-treated cells as evidenced by TUNEL staining, a cell death detection ELISA, and cell viability. Levels of downstream apoptotic mediators, caspase-3 and caspase-9, were significantly (p < 0.05) upregulated following H2O2 treatment but were abrogated following FGF-8 application. Expression levels of Forkhead box protein O1 (FoxO-1), MnSOD, catalase, pAKT, and p-mTOR were significantly (p < 0.05) reduced in the H2O2 group (p < 0.05). Notably, these levels were significantly (p < 0.05) reversed following FGF-8 treatment. Our data, for the first time, suggest FGF-8 is an anti-apoptotic mediator in oxidative-stressed H9c2 cells. Furthermore, our data demonstrate that apoptotic inhibition by FGF-8 is consequent to FoxO-1 oxidative detoxification as well as augmentation to the PI3K/AKT cell survival pathway.


Subject(s)
Apoptosis/drug effects , Fibroblast Growth Factor 8/pharmacology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Animals , Cell Line , Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Nerve Tissue Proteins/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats
5.
Appl Microbiol Biotechnol ; 100(2): 625-35, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26411459

ABSTRACT

Human fibroblast growth factor 8b (FGF8b) was expressed based on a baculovirus expression vector system (BEVS) and identified as having a protective effect on Parkinson's disease. Immunoblotting demonstrated that rhFGF8b proteins were recognized by a human anti-FGF8b antibody. The multiplicity of infection and timing of harvest had a significant effect on protein yield and protein quality. Our results indicated that the rhFGF8b was first detectable at 36 h postinfection and reached a maximum at 60 h. A multiplicity of infection (MOI) of 8 pfu/mL was suitable for harvest. The target protein was purified by heparin-affinity chromatography. In vitro methylthiazol tetrazolium (MTT) assays demonstrated that the purified rhFGF8b could significantly stimulate proliferation of NIH3T3 cells. Furthermore, to elucidate the effect of rhFGF8b on Parkinson's disease, we used FGF8b pretreatment on a cell model of Parkinson's disease. The results indicated that rhFGF8b prevented necrosis and apoptosis of 1-METHYL-4-phenyl pyridine (MPP(+)) treated PC12 cells. Moreover, the effect of FGF8b on messenger RNA (mRNA) levels of apoptosis and ERS genes was investigated to clarify the molecular mechanisms of FGF8b. The results suggest that FGF8b exerts neuroprotective effects by alleviating endoplasmic reticulum (ER) stress during PD. These results suggest that FGF8b may be a promising candidate therapeutic drug for neurodegenerative diseases related to ER stress.


Subject(s)
Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/pharmacology , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Baculoviridae/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromatography, Affinity , Endoplasmic Reticulum Stress/drug effects , Fibroblast Growth Factor 8/biosynthesis , Fibroblast Growth Factor 8/isolation & purification , Humans , Mice , NIH 3T3 Cells , Neuroprotective Agents/isolation & purification , PC12 Cells , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Tetrazolium Salts , Thiazoles
6.
Dev Growth Differ ; 57(6): 421-429, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26100345

ABSTRACT

Limb regeneration ability, which can be observed in amphibians, has been investigated as a representative phenomenon of organ regeneration. Recently, an alternative experimental system called the accessory limb model was developed to investigate early regulation of amphibian limb regeneration. The accessory limb model contributed to identification of limb regeneration inducers in urodele amphibians. Furthermore, the accessory limb model may be applied to other species to explore universality of regeneration mechanisms. This review aims to connect the insights recently gained to emboss universality of regeneration mechanisms among species. The defined molecules (BMP7 (or2) + FGF2 + FGF8) can transform skin wound healing to organ (limb) regeneration responses. The same molecules can initiate regeneration responses in some species.


Subject(s)
Bone Morphogenetic Protein 7/pharmacology , Extremities/physiology , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 8/pharmacology , Regeneration/drug effects , Ambystoma mexicanum/physiology , Amphibians/physiology , Animals , Extremities/growth & development , Regeneration/physiology , Wound Healing/drug effects , Wound Healing/physiology , Xenopus/physiology
7.
Proc Natl Acad Sci U S A ; 108(23): 9703-8, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21606375

ABSTRACT

During early development, midbrain dopaminergic (mDA) neuronal progenitors (NPs) arise from the ventral mesencephalic area by the combined actions of secreted factors and their downstream transcription factors. These mDA NPs proliferate, migrate to their final destinations, and develop into mature mDA neurons in the substantia nigra and the ventral tegmental area. Here, we show that such authentic mDA NPs can be efficiently isolated from differentiated ES cells (ESCs) using a FACS method combining two markers, Otx2 and Corin. Purified Otx2(+)Corin(+) cells coexpressed other mDA NP markers, including FoxA2, Lmx1b, and Glast. Using optimized culture conditions, these mDA NPs continuously proliferated up to 4 wk with almost 1,000-fold expansion without significant changes in their phenotype. Furthermore, upon differentiation, Otx2(+)Corin(+) cells efficiently generated mDA neurons, as evidenced by coexpression of mDA neuronal markers (e.g., TH, Pitx3, Nurr1, and Lmx1b) and physiological functions (e.g., efficient DA secretion and uptake). Notably, these mDA NPs differentiated into a relatively homogenous DA population with few serotonergic neurons. When transplanted into PD model animals, aphakia mice, and 6-OHDA-lesioned rats, mDA NPs differentiated into mDA neurons in vivo and generated well-integrated DA grafts, resulting in significant improvement in motor dysfunctions without tumor formation. Furthermore, grafted Otx2(+)Corin(+) cells exhibited significant migratory function in the host striatum, reaching >3.3 mm length in the entire striatum. We propose that functional and expandable mDA NPs can be efficiently isolated by this unique strategy and will serve as useful tools in regenerative medicine, bioassay, and drug screening.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Mesencephalon/cytology , Neural Stem Cells/cytology , Animals , Cell Line , Cell Proliferation , Dopamine/metabolism , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 8/pharmacology , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Male , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Motor Activity , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/physiopathology , Parkinson Disease, Secondary/surgery , Rats , Rats, Sprague-Dawley , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Stem Cell Transplantation/methods
8.
Int Endod J ; 47(4): 346-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24033427

ABSTRACT

AIM: The aim of this study was to differentiate human embryonic stem cells (hESCs) into odontoblastic lineage in an optimized culture milieu. METHODOLOGY: In Phase 1, hESCs were differentiated into mesenchymal stem cells (H9-MSCs). In Phase 2, H9-MSCs were then differentiated into odontoblast-like cells (H9-Odont) under the stimulation of FGF-8 and BMP-4. Alternatively, H9-MSCs were differentiated into osteogenic lineage (H9-Osteo). In Phase 3, H9-Odont were seeded on 17% EDTA-treated dentine substrates in the presence of FGF-8 and BMP-4 for further differentiation. All experiments were performed in triplicate (n = 3). One-way anova was used to test hESC differentiation into different cell types. Post hoc Tukey's test was used to compare between groups. P < 0.05 was considered statistically significant. RESULTS: H9-Odont expressed the odontoblastic marker DSPP gene 125.47 ± 0.1 (SD)-folds higher compared with H9-MSCs at mRNA level (real-time RT-PCR). Additionally, the flow cytometry results revealed 53.1 ± 3.4 (SD) % of DSP (+) cells in H9-Odont. Alternatively, H9-Osteo expressed 5.9 ± 2.2 (SD) % of DSP (+) cells. Moreover, the SEM results demonstrated that H9-Odont were found to undergo morphological changes from a fibroblast-like shape into more rounded shapes with cytoplasmic extensions into the dentinal tubules when seeded on 17% EDTA-treated dentine substrate in the presence of FGF-8 and BMP-4. However, H9-Osteo and H9-MSCs did not show similar morphological changes under similar culture milieu. CONCLUSION: This study supports the potential of hESCs as a stable, consistent, unlimited and 'off-the-shelf' cell source to obtain odontoblastic cells for future clinical and research applications.


Subject(s)
Cell Differentiation/physiology , Human Embryonic Stem Cells/cytology , Odontoblasts/cytology , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Fibroblast Growth Factor 8/pharmacology , Flow Cytometry , Humans , In Vitro Techniques , Microscopy, Electron, Scanning , Phenotype , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
J Neurosci ; 32(22): 7477-92, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22649227

ABSTRACT

Adult zebrafish show a remarkable capacity to regenerate their spinal column after injury, an ability that stands in stark contrast to the limited repair that occurs within the mammalian CNS post-injury. The reasons for this interspecies difference in regenerative capacity remain unclear. Here we demonstrate a novel role for Fgf signaling during glial cell morphogenesis in promoting axonal regeneration after spinal cord injury. Zebrafish glia are induced by Fgf signaling, to form an elongated bipolar morphology that forms a bridge between the two sides of the resected spinal cord, over which regenerating axons actively migrate. Loss of Fgf function inhibits formation of this "glial bridge" and prevents axon regeneration. Despite the poor potential for mammalian axonal regeneration, primate astrocytes activated by Fgf signaling adopt a similar morphology to that induced in zebrafish glia. This suggests that differential Fgf regulation, rather than intrinsic cell differences, underlie the distinct responses of mammalian and zebrafish glia to injury.


Subject(s)
Nerve Regeneration/physiology , Neuroglia/physiology , Signal Transduction/genetics , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Analysis of Variance , Animals , Animals, Genetically Modified , Bromodeoxyuridine/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Dextrans , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 3/genetics , Fibroblast Growth Factor 3/metabolism , Fibroblast Growth Factor 8/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/genetics , Green Fluorescent Proteins/genetics , Humans , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Ki-67 Antigen/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Motor Activity/drug effects , Motor Activity/genetics , Nerve Regeneration/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neuroglia/drug effects , Pyrroles/pharmacology , RNA, Messenger , Receptor, Fibroblast Growth Factor, Type 1/genetics , Recovery of Function , Rhodamines , Signal Transduction/drug effects , Time Factors , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
Dev Biol ; 349(2): 137-46, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20969842

ABSTRACT

Despite their importance to oral health, the mechanisms of minor salivary gland (SG) development are largely unexplored. Here we present in vivo and in vitro analyses of developing minor SGs in wild type and mutant mice. Eda, Shh and Fgf signalling pathway genes are expressed in these glands from an early stage of development. Developing minor SGs are absent in Eda pathway mutant embryos, and these mice exhibit a dysplastic circumvallate papilla with disrupted Shh expression. Supplementation of Eda pathway mutant minor SG explants with recombinant EDA rescues minor SG induction. Supplementation with Fgf8 or Shh, previously reported targets of Eda signalling, leads to induction of gland like structures in a few cases, but these fail to develop into minor SGs.


Subject(s)
Ectodysplasins/metabolism , Recombinant Proteins/pharmacology , Salivary Glands, Minor/embryology , Signal Transduction/physiology , Animals , DNA Primers/genetics , Ectodysplasins/genetics , Fibroblast Growth Factor 8/metabolism , Fibroblast Growth Factor 8/pharmacology , Genotype , Hedgehog Proteins/metabolism , Hedgehog Proteins/pharmacology , Histological Techniques , In Situ Hybridization , Mice , Mice, Mutant Strains , Polymerase Chain Reaction , Recombinant Proteins/metabolism , Salivary Glands, Minor/drug effects
11.
Dev Biol ; 354(1): 18-30, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21419761

ABSTRACT

Cardiac neural crest cells migrate into the pharyngeal arches where they support development of the pharyngeal arch arteries. The pharyngeal endoderm and ectoderm both express high levels of FGF8. We hypothesized that FGF8 is chemotactic for cardiac crest cells. To begin testing this hypothesis, cardiac crest was explanted for migration assays under various conditions. Cardiac neural crest cells migrated more in response to FGF8. Single cell tracing indicated that this was not due to proliferation and subsequent transwell assays showed that the cells migrate toward an FGF8 source. The migratory response was mediated by FGF receptors (FGFR) 1 and 3 and MAPK/ERK intracellular signaling. To test whether FGF8 is chemokinetic and/or chemotactic in vivo, dominant negative FGFR1 was electroporated into the premigratory cardiac neural crest. Cells expressing the dominant negative receptor migrated slower than normal cardiac neural crest cells and were prone to remain in the vicinity of the neural tube and die. Treating with the FGFR1 inhibitor, SU5402 or an FGFR3 function-blocking antibody also slowed neural crest migration. FGF8 over-signaling enhanced neural crest migration. Neural crest cells migrated to an FGF8-soaked bead placed dorsal to the pharynx. Finally, an FGF8 producing plasmid was electroporated into an ectopic site in the ventral pharyngeal endoderm. The FGF8 producing cells attracted a thick layer of mesenchymal cells. DiI labeling of the neural crest as well as quail-to-chick neural crest chimeras showed that neural crest cells migrated to and around the ectopic site of FGF8 expression. These results showing that FGF8 is chemotactic and chemokinetic for cardiac neural crest adds another dimension to understanding the relationship of FGF8 and cardiac neural crest in cardiovascular defects.


Subject(s)
Cell Movement/drug effects , Chemotaxis/drug effects , Fibroblast Growth Factor 8/pharmacology , Neural Crest/cytology , Animals , Apoptosis/drug effects , Butadienes/pharmacology , Cell Proliferation/drug effects , Chick Embryo , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart/embryology , Immunohistochemistry , In Situ Hybridization , Mesoderm/embryology , Mesoderm/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Myocardium/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Nitriles/pharmacology , Pharynx/embryology , Pharynx/metabolism , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction
12.
Brain ; 134(Pt 6): 1808-28, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21576111

ABSTRACT

Senataxin is encoded by the SETX gene and is mainly involved in two different neurodegenerative diseases, the dominant juvenile form of amyotrophic lateral sclerosis type 4 and a recessive form of ataxia with oculomotor apraxia type 2. Based on protein homology, senataxin is predicted to be a putative DNA/RNA helicase, while senataxin interactors from patients' lymphoblast cell lines suggest a possible involvement of the protein in different aspects of RNA metabolism. Except for an increased sensitivity to oxidative DNA damaging agents shown by some ataxia with neuropathy patients' cell lines, no data are available about possible functional consequences of dominant SETX mutations and no studies address the function of senataxin in neurons. To start elucidating the physiological role of senataxin in neurons and how disease-causing mutations in this protein lead to neurodegeneration, we analysed the effect of senataxin on neuronal differentiation in primary hippocampal neurons and retinoic acid-treated P19 cells by modulating the expression levels of wild-type senataxin and three different dominant mutant forms of the protein. Wild-type senataxin overexpression was required and sufficient to trigger neuritogenesis and protect cells from apoptosis during differentiation. These actions were reversed by silencing of senataxin. In contrast, overexpression of the dominant mutant forms did not affect the regular differentiation process in primary hippocampal neurons. Analysis of the cellular pathways leading to neuritogenesis and cytoprotection revealed a role of senataxin in modulating the expression levels and signalling activity of fibroblast growth factor 8. Silencing of senataxin reduced, while overexpression enhanced, fibroblast growth factor 8 expression levels and the phosphorylation of related target kinases and effector proteins. The effects of senataxin overexpression were prevented when fibroblast growth factor 8 signalling was inhibited, while exogenous fibroblast growth factor 8 reversed the effects of senataxin silencing. Overall, these results reveal a key role of senataxin in neuronal differentiation through the fibroblast growth factor 8 signalling and provide initial molecular bases to explain the neurodegeneration associated with loss-of-function mutations in senataxin found in recessive ataxia. The lack of effect on neuritogenesis observed with the overexpression of the dominant mutant forms of senataxin apparently excludes a dominant negative effect of these mutants while favouring haploinsufficiency as the pathogenic mechanism implicated in the amyotrophic lateral sclerosis 4-related degenerative condition. Alternatively, a different protein function, other than the one involved in neuritogenesis, may be implicated in these dominant degenerative processes.


Subject(s)
DNA Helicases/metabolism , Fibroblast Growth Factor 8/pharmacology , Neurites/drug effects , Neurites/physiology , Neurons/cytology , RNA Helicases/metabolism , Signal Transduction/physiology , Animals , Caspase 3/metabolism , Cell Death/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , DNA Helicases/genetics , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Humans , Mice , Multifunctional Enzymes , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroblastoma/pathology , Neurons/drug effects , RNA Helicases/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Time Factors , Transfection/methods , Tretinoin/pharmacology
13.
Dev Dyn ; 240(6): 1344-53, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21412937

ABSTRACT

Regression of vestigial tooth buds results in the formation of the toothless diastema, a unique feature of the mouse dentition. Revitalization of the diastemal vestigial tooth bud provides an excellent model for studying tooth regeneration and replacement. It has been previously shown that suppression of fibroblast growth factor (FGF) signaling in the diastema results in vestigial tooth bud regression. In this study, we report that application of exogenous FGF8 to the mouse embryonic diastemal region rescues diastemal tooth development. However, this rescue of diastemal tooth development occurs only in an isolated diastemal regions and not in the mandibular quadrant, which includes the incisor and molar germs. FGF8 promotes cell proliferation and inhibits apoptosis in diastemal tooth epithelium, and revitalizes the tooth developmental program, as evidenced by the expression of genes critical for normal tooth development. Our results also support the idea that the adjacent tooth germs contribute to the suppression of diastemal vestigial tooth buds by means of multiple signals.


Subject(s)
Diastema/embryology , Fibroblast Growth Factor 8/pharmacology , Tooth/drug effects , Tooth/growth & development , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Cell Proliferation/drug effects , Cells, Cultured , Female , Gene Expression Regulation, Developmental/drug effects , Male , Mice , Odontogenesis/drug effects , Odontogenesis/genetics , Pregnancy , Tooth/metabolism , Tooth Germ/drug effects , Tooth Germ/embryology
14.
Dis Model Mech ; 15(8)2022 08 01.
Article in English | MEDLINE | ID: mdl-35833364

ABSTRACT

Fibroblast growth factor 8 (FGF8), acting through the fibroblast growth factor receptor 1 (FGFR1), has an important role in the development of gonadotropin-releasing hormone-expressing neurons (GnRH neurons). We hypothesized that FGF8 regulates differentiation of human GnRH neurons in a time- and dose-dependent manner via FGFR1. To investigate this further, human pluripotent stem cells were differentiated during 10 days of dual-SMAD inhibition into neural progenitor cells, followed either by treatment with FGF8 at different concentrations (25 ng/ml, 50 ng/ml or 100 ng/ml) for 10 days or by treatment with 100 ng/ml FGF8 for different durations (2, 4, 6 or 10 days); cells were then matured through DAPT-induced inhibition of Notch signaling for 5 days into GnRH neurons. FGF8 induced expression of GNRH1 in a dose-dependent fashion and the duration of FGF8 exposure correlated positively with gene expression of GNRH1 (P<0.05, Rs=0.49). However, cells treated with 100 ng/ml FGF8 for 2 days induced the expression of genes, such as FOXG1, ETV5 and SPRY2, and continued FGF8 treatment induced the dynamic expression of several other genes. Moreover, during exposure to FGF8, FGFR1 localized to the cell surface and its specific inhibition with the FGFR1 inhibitor PD166866 reduced expression of GNRH1 (P<0.05). In neurons, FGFR1 also localized to the nucleus. Our results suggest that dose- and time-dependent FGF8 signaling via FGFR1 is indispensable for human GnRH neuron ontogeny. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Gonadotropin-Releasing Hormone , Receptor, Fibroblast Growth Factor, Type 1 , Fibroblast Growth Factor 8/metabolism , Fibroblast Growth Factor 8/pharmacology , Forkhead Transcription Factors/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism
15.
BMC Neurosci ; 12: 25, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21385431

ABSTRACT

BACKGROUND: Deletion or mutation(s) of the survival motor neuron 1 (SMN1) gene causes spinal muscular atrophy (SMA), a neuromuscular disease characterized by spinal motor neuron death and muscle paralysis. Complete loss of the SMN protein is embryonically lethal, yet reduced levels of this protein result in selective death of motor neurons. Why motor neurons are specifically targeted by SMN deficiency remains to be determined. In this study, embryonic stem (ES) cells derived from a severe SMA mouse model were differentiated into motor neurons in vitro by addition of retinoic acid and sonic hedgehog agonist. Proteomic and western blot analyses were used to probe protein expression alterations in this cell-culture model of SMA that could be relevant to the disease. RESULTS: When ES cells were primed with Noggin/fibroblast growth factors (bFGF and FGF-8) in a more robust neural differentiation medium for 2 days before differentiation induction, the efficiency of in vitro motor neuron differentiation was improved from ~25% to ~50%. The differentiated ES cells expressed a pan-neuronal marker (neurofilament) and motor neuron markers (Hb9, Islet-1, and ChAT). Even though SMN-deficient ES cells had marked reduced levels of SMN (~20% of that in control ES cells), the morphology and differentiation efficiency for these cells are comparable to those for control samples. However, proteomics in conjunction with western blot analyses revealed 6 down-regulated and 14 up-regulated proteins with most of them involved in energy metabolism, cell stress-response, protein degradation, and cytoskeleton stability. Some of these activated cellular pathways showed specificity for either undifferentiated or differentiated cells. Increased p21 protein expression indicated that SMA ES cells were responding to cellular stress. Up-regulation of p21 was confirmed in spinal cord tissues from the same SMA mouse model from which the ES cells were derived. CONCLUSION: SMN-deficient ES cells provide a cell-culture model for SMA. SMN deficiency activates cellular stress pathways, causing a dysregulation of energy metabolism, protein degradation, and cytoskeleton stability.


Subject(s)
Gene Expression Regulation/physiology , Motor Neurons/metabolism , Muscular Atrophy, Spinal/pathology , Proteome/metabolism , Proteomics/methods , Spinal Cord/pathology , Animals , Antibodies/pharmacology , Cell Differentiation/drug effects , Cell Line, Transformed , Disease Models, Animal , Embryo, Mammalian , Fibroblast Growth Factor 8/pharmacology , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Mice , Mice, Transgenic , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proteome/analysis , Proteome/immunology , Stem Cells/drug effects , Stem Cells/metabolism , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism
16.
Stem Cells ; 28(10): 1772-81, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20715182

ABSTRACT

Formation of the neural plate is an intricate process in early mammalian embryonic development mediated by cells of the inner cell mass and involving a series of steps, including development of the epiblast. Here, we report on the creation of an embryonic stem (ES) cell-based system to isolate and identify neural induction intermediates with characteristics of epiblast cells and neural plate. We demonstrate that neural commitment requires prior differentiation of ES cells into epiblast cells that are indistinguishable from those derived from natural embryos. We also demonstrate that epiblast cells can be isolated and cultured as epiblast stem cell lines. Fgf signaling is shown to be required for the differentiation of ES cells into these epiblast cells. Fgf2, widely used for maintenance of both human ES cells and epiblast stem cells, inhibits formation of early neural cells by epiblast intermediates in a dose-dependent manner and is sufficient to promote transient self-renewal of epiblast stem cells. In contrast, Fgf8, the endogenous embryonic neural inducer, fails to promote epiblast self-renewal, but rather promotes more homogenous neural induction with transient self-renewal of early neural cells. Removal of Fgf signaling entirely from epiblast cells promotes rapid neural induction and subsequent neurogenesis. We conclude that Fgf signaling plays different roles during the differentiation of ES cells, with an initial requirement in epiblast formation and a subsequent role in self-renewal. Fgf2 and Fgf8 thus stimulate self-renewal in different cell types.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblast Growth Factors/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 8/pharmacology , Germ Layers/cytology , Germ Layers/drug effects , Humans , Male , Mice , Neural Plate/cytology , Neural Plate/drug effects , Neurogenesis/drug effects , Neurogenesis/genetics , Signal Transduction/drug effects
17.
Exp Cell Res ; 316(7): 1148-58, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20171210

ABSTRACT

In this study we demonstrated that neural rosettes derived from human ES cells can give rise either to neural crest precursors, following expansion in presence of bFGF and EGF, or to dopaminergic precursors after exposure to ventralizing factors Shh and FGF8. Both regionalised precursors are capable of extensive proliferation and differentiation towards the corresponding terminally differentiated cell types. In particular, peripheral neurons, cartilage, bone, smooth muscle cells and also pigmented cells were obtained from neural crest precursors while tyrosine hydroxylase and Nurr1 positive dopaminergic neurons were derived from FGF8 and Shh primed rosette cells. Gene expression and immunocytochemistry analyses confirmed the expression of dorsal and neural crest genes such as Sox10, Slug, p75, FoxD3, Pax7 in neural precursors from bFGF-EGF exposed rosettes. By contrast, priming of rosettes with FGF8 and Shh induced the expression of dopaminergic markers Engrailed1, Pax2, Pitx3, floor plate marker FoxA2 and radial glia markers Blbp and Glast, the latter in agreement with the origin of dopaminergic precursors from floor plate radial glia. Moreover, in vivo transplant of proliferating Shh/FGF8 primed precursors in parkinsonian rats demonstrated engraftment and terminal dopaminergic differentiation. In conclusion, we demonstrated the derivation of long-term self-renewing precursors of selected regional identity as potential cell reservoirs for cell therapy applications, such as CNS degenerative diseases, or for the development of toxicological tests.


Subject(s)
Cell Differentiation/drug effects , Central Nervous System/drug effects , Fibroblast Growth Factor 8/pharmacology , Hedgehog Proteins/pharmacology , Neural Plate/drug effects , Stem Cells/drug effects , Animals , Body Patterning/drug effects , Cell Culture Techniques , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Central Nervous System/embryology , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Neural Plate/embryology , Neurons/drug effects , Neurons/physiology , Neurons/transplantation , Rats , Rats, Sprague-Dawley , Spheroids, Cellular/drug effects , Spheroids, Cellular/physiology , Spheroids, Cellular/transplantation , Stem Cells/physiology , Time Factors , Transplantation, Heterologous
18.
Mol Cell Neurosci ; 45(3): 258-66, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20603216

ABSTRACT

The cardinal motor symptoms of Parkinson's disease (PD) are caused by the vulnerability to dysfunction and degeneration of ventral midbrain (VM) dopaminergic (DA) neurons. A major limitation for experimental studies of current ES/iPS cell differentiation protocols is the lack of VM DA neurons with a stable phenotype as defined by an expression marker code of FOXA2/TH/ß-tubulin. Here we demonstrate a combination of three modifications that were required to produce VM DA neurons. Firstly, early and specific exposure to 10(-)(8)M (low dose) retinoic acid improved the regional identity of neural progenitor cells derived from human ES cells, PD or healthy subject-specific iPS cells. Secondly, a high activity form of human sonic hedgehog established a sizeable FOXA2(+) neural progenitor cell population in vitro. Thirdly, early exposure to FGF8a, rather than Fgf8b, and WNT1 was required for robust differentiation of the FOXA2(+) floor plate-like human neural progenitor cells into FOXA2(+) DA neurons. FOXA2(+) DA neurons were also generated when this protocol was adapted to feeder-free conditions. In summary, this new human ES and iPS cell differentiation protocol using FGF8a, WNT1, low dose retinoic acid and a high activity form of SHH can generate human VM DA neurons that are required for relevant new bioassays, drug discovery and cell based therapies for PD.


Subject(s)
Cell Differentiation/drug effects , Dopamine/metabolism , Embryonic Stem Cells/cytology , Fibroblast Growth Factor 8/pharmacology , Hedgehog Proteins/metabolism , Neurons/cytology , Pluripotent Stem Cells/cytology , Tretinoin/pharmacology , Animals , Cell Culture Techniques , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Hedgehog Proteins/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Mesencephalon/cytology , Mice , Neurons/drug effects , Neurons/metabolism , Parkinson Disease , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/physiology , Wnt1 Protein/pharmacology
19.
Endocrinology ; 162(9)2021 09 01.
Article in English | MEDLINE | ID: mdl-34125902

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus play a key role in the regulation of reproductive function. In this study, we sought an efficient method for generating GnRH neurons from human embryonic and induced pluripotent stem cells (hESC and hiPSC, respectively). First, we found that exposure of primitive neuroepithelial cells, rather than neuroprogenitor cells, to fibroblast growth factor 8 (FGF8), was more effective in generating GnRH neurons. Second, addition of kisspeptin to FGF8 further increased the efficiency rates of GnRH neurogeneration. Third, we generated a fluorescent marker mCherry labeled human embryonic GnRH cell line (mCh-hESC) using a CRISPR-Cas9 targeting approach. Fourth, we examined physiological characteristics of GnRH (mCh-hESC) neurons: similar to GnRH neurons in vivo, they released the GnRH peptide in a pulsatile manner at ~60 min intervals; GnRH release increased in response to high potassium, kisspeptin, estradiol, and neurokinin B challenges; and injection of depolarizing current induced action potentials. Finally, we characterized developmental changes in transcriptomes of GnRH neurons using hESC, hiPSC, and mCh-hESC. The developmental pattern of transcriptomes was remarkably similar among the 3 cell lines. Collectively, human stem cell-derived GnRH neurons will be an important tool for establishing disease models to understand diseases, such as idiopathic hypothalamic hypogonadism, and testing contraceptive drugs.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Human Embryonic Stem Cells/physiology , Neurogenesis/genetics , Neurons/physiology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Fibroblast Growth Factor 8/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Human Embryonic Stem Cells/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Transcriptome/drug effects
20.
J Neurosci ; 29(13): 4044-55, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19339600

ABSTRACT

Accumulating evidence indicates that signaling centers controlling the dorsoventral (DV) polarization of the neural tube, the roof plate and the floor plate, play crucial roles in axon guidance along the DV axis. However, the role of signaling centers regulating the rostrocaudal (RC) polarization of the neural tube in axon guidance along the RC axis remains unknown. Here, we show that a signaling center located at the midbrain-hindbrain boundary (MHB) regulates the rostrally directed growth of axons from midbrain dopaminergic neurons (mDANs). We found that beads soaked with fibroblast growth factor 8 (FGF8), a signaling molecule that mediates patterning activities of the MHB, repelled mDAN axons that extended through the diencephalon. This repulsion may be mediated by semaphorin 3F (sema3F) because (1) FGF8-soaked beads induced an increase in expression of sema3F, (2) sema3F expression in the midbrain was essentially abolished by the application of an FGF receptor tyrosine kinase inhibitor, and (3) mDAN axonal growth was also inhibited by sema3F. Furthermore, mDAN axons expressed a sema3F receptor, neuropilin-2 (nrp2), and the removal of nrp-2 by gene targeting caused caudal growth of mDAN axons. These results indicate that the MHB signaling center regulates the growth polarity of mDAN axons along the RC axis by inducing sema3F.


Subject(s)
Axons/drug effects , Dopamine/metabolism , Fibroblast Growth Factor 8/pharmacology , Mesencephalon/cytology , Neurons/cytology , Semaphorins/metabolism , Age Factors , Animals , Animals, Newborn , Axons/physiology , Body Patterning/drug effects , Body Patterning/genetics , Brain/embryology , Brain/growth & development , Brain/metabolism , COS Cells , Chlorocebus aethiops , Coculture Techniques/methods , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Electroporation/methods , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Mesencephalon/embryology , Mesencephalon/growth & development , Mice , Mice, Knockout , Neurons/drug effects , Neuropilin-2/deficiency , Neuropilin-2/metabolism , Organ Culture Techniques , Otx Transcription Factors/metabolism , Pyrroles/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL