Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Bull Exp Biol Med ; 176(5): 576-580, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38724808

ABSTRACT

We performed a comparative in vitro study of the involvement of NF-κB, PI3K, cAMP, ERK1/2, p38, JAKs, STAT3, JNK, and p53-dependent intracellular signaling in the functioning of neural stem cells (NSC) under the influence of basic fibroblast growth factor (FGF) and FGF receptor agonist, diterpene alkaloid songorine. The significant differences in FGFR-mediated intracellular signaling in NSC were revealed for these ligands. In both cases, stimulation of progenitor cell proliferation occurs with the participation of NF-κB, PI3K, ERK1/2, JAKs, and STAT3, while JNK and p53, on the contrary, inhibit cell cycle progression. However, under the influence of songorin, cAMP- and p38-mediated cascades are additionally involved in the transmission of the NSC division-activating signal. In addition, unlike FGF, the alkaloid stimulates progenitor cell differentiation by activating ERK1/2, p38, JNK, p53, and STAT3.


Subject(s)
Cell Differentiation , Cell Proliferation , Diterpenes , Neural Stem Cells , Receptors, Fibroblast Growth Factor , STAT3 Transcription Factor , Signal Transduction , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Animals , STAT3 Transcription Factor/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Fibroblast Growth Factor/agonists , Signal Transduction/drug effects , Cell Proliferation/drug effects , Diterpenes/pharmacology , Cell Differentiation/drug effects , NF-kappa B/metabolism , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/agonists , Phosphatidylinositol 3-Kinases/metabolism , Alkaloids/pharmacology , MAP Kinase Signaling System/drug effects , Janus Kinases/metabolism , Cyclic AMP/metabolism , Cells, Cultured , Rats
2.
Methods Mol Biol ; 2312: 301-305, 2021.
Article in English | MEDLINE | ID: mdl-34228298

ABSTRACT

Cell culture media are often supplemented with recombinant growth factors and cytokines to reproduce biological conditions in vitro. Basic fibroblast growth factor (bFGF) has been widely used to support the pluripotency and self-renewal activity of human induced pluripotent stem cells (hiPSCs). We had previously developed a synthetic surrogate for bFGF on the basis of a DNA aptamer that binds to one of the FGF receptors. Since DNA aptamers have advantages over recombinant proteins in terms of thermal stability and production cost, replacing recombinant growth factors in cell culture media with DNA aptamers would be of great interest. Herein, we describe our protocol for feeder-free hiPSC culture using a DNA aptamer-based mimic of bFGF.


Subject(s)
Aptamers, Nucleotide/pharmacology , Fibroblast Growth Factor 2/pharmacology , Induced Pluripotent Stem Cells/drug effects , Molecular Mimicry , Receptors, Fibroblast Growth Factor/agonists , Cell Culture Techniques , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Cells, Cultured , Culture Media , Humans , Induced Pluripotent Stem Cells/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction
3.
J Cell Physiol ; 236(11): 7642-7654, 2021 11.
Article in English | MEDLINE | ID: mdl-33959949

ABSTRACT

Multiple paracrine factors regulate the barrier properties of human brain capillary endothelial cells (BCECs). Understanding the precise mode of action of these factors remains a challenging task, because of the limited availability of functionally competent BCECs and the use of serum-containing medium. In the present study, we employed a defined protocol for producing BCECs from human inducible pluripotent stem cells. We found that autocrine secretion of basic fibroblast growth factor (bFGF) is necessary for the establishment a tight BCECs barrier, as revealed by measurements of transendothelial electric resistance (TEER). In contrast, addition of exogenous bFGF in concentrations higher than 4 ng/ml inhibited TEER in a concentration-dependent manner. Exogenous bFGF did not significantly affect expression and distribution of tight junction proteins claudin-5, occludin and zonula occludens (ZO)-1. Treatment with FGF receptor blocker PD173074 (15 µM) suppressed inhibitory effects of bFGF and induced nuclear translocation of protein ZO-1. Inhibition of phosphoinositide 3-Kinase (PI-3K) with LY294002 (25 µM) significantly potentiated an inhibitory effect of bFGF on TEER indicating that PI-3K signalling pathway counteracts bFGF modulation of TEER. In conclusion, we show that autocrine bFGF secretion is necessary for the proper barrier function of BCECs, whereas exogenous bFGF in higher doses suppresses barrier resistance. Our findings demonstrate a dual role for bFGF in the regulation of BCEC barrier function.


Subject(s)
Brain/blood supply , Capillaries/drug effects , Endothelial Cells/drug effects , Fibroblast Growth Factor 2/pharmacology , Induced Pluripotent Stem Cells/drug effects , Autocrine Communication , Capillaries/metabolism , Cell Differentiation , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Electric Impedance , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Permeability , Phosphatidylinositol 3-Kinase/metabolism , Receptors, Fibroblast Growth Factor/agonists , Receptors, Fibroblast Growth Factor/metabolism
4.
J Cosmet Dermatol ; 19(2): 477-484, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31099492

ABSTRACT

BACKGROUND: Fibroblast growth factors (FGFs) are promising agents with which to treat problems of skin and hair. But their inability to penetrate into the skin due to their large size and hydrophilic nature prevents their topical application as effective cosmetic ingredients. AIMS: To identify small peptide(s) with FGF-like activity and epidermis permeability. METHODS: Several peptides deduced from our earlier studies were tested for their ability to promote keratinocyte growth and to activate FGF receptors (FGFRs). Permeability was assessed using HPLC after derivatization. RESULTS: A dipeptide, prolyl-isoleucine (Pro-Ile), not only stimulated growth of human keratinocytes, it also moderately activated FGFR3c and FGFR4, and activated FGFR1c to a lesser extent. This receptor specificity of Pro-Ile is similar to that of FGF18. The activity of Pro-Ile toward FGFR/BaF3 cells was enhanced by heparin and was inhibited by an FGFR inhibitor, PD173074. Pro-Ile enhanced the activity of 5 ng/mL FGF18, but suppressed the activity of 50 ng/mL FGF18 toward FGFR3c and FGFR4. Pro-Ile was found to permeate through validated model human epidermis. CONCLUSIONS: These results indicate that the dipeptide Pro-Ile acts as a partial agonist/antagonist for FGFR signaling, that it has receptor specificity similar to FGF18, and that it is able to penetrate into the model epidermis. Because FGFs expressed in the cutaneous system are physiological regulators, these results suggest the potential utility of this peptide as a topically applicable cosmetic ingredient for the regulation of skin physiology, hair growth, and wound healing.


Subject(s)
Cosmetics/pharmacology , Dipeptides/pharmacology , Epidermis/metabolism , Receptors, Fibroblast Growth Factor/agonists , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Cell Line , Humans , Keratinocytes , Mice , Permeability , Signal Transduction
5.
Curr Drug Targets ; 21(5): 445-457, 2020.
Article in English | MEDLINE | ID: mdl-31670620

ABSTRACT

AIMS: The aim of this study is to provide an overview of several emerging anti-diabetic molecules. BACKGROUND: Diabetes is a complex metabolic disorder involving the dysregulation of glucose homeostasis at various levels. Insulin, which is produced by ß-pancreatic cells, is a chief regulator of glucose metabolism, regulating its consumption within cells, which leads to energy generation or storage as glycogen. Abnormally low insulin secretion from ß-cells, insulin insensitivity, and insulin tolerance lead to higher plasma glucose levels, resulting in metabolic complications. The last century has witnessed extraordinary efforts by the scientific community to develop anti-diabetic drugs, and these efforts have resulted in the discovery of exogenous insulin and various classes of oral anti-diabetic drugs. OBJECTIVE: Despite these exhaustive anti-diabetic pharmaceutical and therapeutic efforts, long-term glycemic control, hypoglycemic crisis, safety issues, large-scale economic burden and side effects remain the core problems. METHODS: The last decade has witnessed the development of various new classes of anti-diabetic drugs with different pharmacokinetic and pharmacodynamic profiles. Details of their FDA approvals and advantages/disadvantages are summarized in this review. RESULTS: The salient features of insulin degludec, sodium-glucose co-transporter 2 inhibitors, glucokinase activators, fibroblast growth factor 21 receptor agonists, and GLP-1 agonists are discussed. CONCLUSION: In the future, these new anti-diabetic drugs may have broad clinical applicability. Additional multicenter clinical studies on these new drugs should be conducted.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Animals , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Enzyme Activators/pharmacology , Glucagon-Like Peptide 1/analogs & derivatives , Humans , Insulin, Long-Acting/pharmacology , Insulin, Long-Acting/therapeutic use , Receptors, Fibroblast Growth Factor/agonists
6.
J Neurosci ; 36(16): 4534-48, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-27098696

ABSTRACT

Aberrant branch formation of granule cell axons (mossy fiber sprouting) is observed in the dentate gyrus of many patients with temporal lobe epilepsy and in animal models of epilepsy. However, the mechanisms underlying mossy fiber sprouting remain elusive. Based on the hypothesis that seizure-mediated gene expression induces abnormal mossy fiber growth, we screened activity-regulated genes in the hippocampus and found that neuritin, an extracellular protein anchored to the cell surface, was rapidly upregulated after electroconvulsive seizures. Overexpression of neuritin in the cultured rat granule cells promoted their axonal branching. Also, kainic acid-dependent axonal branching was abolished in the cultured granule cells fromneuritinknock-out mice, suggesting that neuritin may be involved in activity-dependent axonal branching. Moreover,neuritinknock-out mice showed less-severe seizures in chemical kindling probably by reduced mossy fiber sprouting and/or increased seizure resistance. We found that inhibition of the fibroblast growth factor (FGF) receptor attenuated the neuritin-dependent axonal branching. FGF administration also increased branching in granule neurons, whereasneuritinknock-out mice did not show FGF-dependent axonal branching. In addition, FGF and neuritin treatment enhanced the recruitment of FGF receptors to the cell surface. These findings suggest that neuritin and FGF cooperate in inducing mossy fiber sprouting through FGF signaling. Together, these results suggest that FGF and neuritin-mediated axonal branch induction are involved in the aggravation of epilepsy. SIGNIFICANCE STATEMENT: This study reveals the molecular mechanism underlying mossy fiber sprouting. Mossy fiber sprouting is the aberrant axonal branching of granule neurons in the hippocampus, which is observed in patients with epilepsy. Excess amounts of neuritin, a protein upregulated by neural activity, promoted axonal branching in granule neurons. A deficiency of neuritin suppressed mossy fiber sprouting and resulted in mitigation of seizure severity. Neuritin and fibroblast growth factor (FGF) cooperated in stimulating FGF signaling and enhancing axonal branching. Neuritin is necessary for FGF-mediated recruitment of FGF receptors to the cell surface. The recruitment of FGF receptors would promote axonal branching. The discovery of this new mechanism should contribute to the development of novel antiepileptic drugs to inhibit axonal branching via neuritin-FGF signaling.


Subject(s)
Axons/metabolism , Fibroblast Growth Factors/metabolism , Nerve Tissue Proteins/physiology , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/physiology , Animals , Axons/drug effects , Female , Fibroblast Growth Factors/pharmacology , GPI-Linked Proteins/pharmacology , GPI-Linked Proteins/physiology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/pharmacology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Receptors, Fibroblast Growth Factor/agonists , Signal Transduction/drug effects
7.
Mol Neurobiol ; 53(1): 584-594, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25502296

ABSTRACT

The CD200 ligand is expressed by a variety of cell types, including vascular endothelia, kidney glomeruli, some subsets of T and B cells, and neurons in the brain and periphery. In contrast, the receptor of CD200, CD200R, has a limited expression pattern and is mainly expressed by cells of myeloid origin. A recently solved crystal structure of the CD200-CD200R ectodomain complex suggests involvement of the first immunoglobulin (Ig)-like modules in ligand-receptor binding, resulting in the inhibition of myeloid cell function. In the central nervous system, CD200 has been implicated in the suppression of microglia activation. We for the first time demonstrated that CD200 can interact with and transduce signaling through activation of the fibroblast growth factor receptor (FGFR), thereby inducing neuritogenesis and promoting neuronal survival in primary neurons. CD200-induced FGFR phosphorylation was abrogated by CD200R, whereas FGF2-induced FGFR activation was inhibited by CD200. We also identified a sequence motif located in the first Ig-like module of CD200, likely representing the minimal CD200 binding site for FGFR. The FGFR binding motif overlaps with the CD200R binding site, suggesting that they can compete for CD200 binding in cells that express both receptors. We propose that CD200 in neurons functions as a ligand of FGFR.


Subject(s)
Antigens, CD/metabolism , Immunologic Factors/metabolism , Nerve Growth Factors/metabolism , Neurons/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Sequence , Animals , Antigens, CD/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Immunologic Factors/pharmacology , Molecular Sequence Data , Nerve Growth Factors/genetics , Neurons/drug effects , Protein Binding/physiology , Protein Structure, Secondary , Rats , Rats, Wistar , Receptors, Fibroblast Growth Factor/agonists , Receptors, Fibroblast Growth Factor/genetics
8.
Reproduction ; 145(2): 191-201, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23241344

ABSTRACT

The overall aim of this work was to examine the expression profiles for fibroblast growth factor receptors (FGFRs) and describe their biological importance during bovine pre- and peri-implantation conceptus development. FGFR1 and FGFR2 mRNAs were detected at 1-, 2-, 8-cell, morula and blastocyst stages whereas FGFR3 and FGFR4 mRNAs were detected after the 8-cell stage but not earlier. The abundance of FGFR1, FGFR3, and FGFR4 mRNAs increased at the morula and blastocyst stages. Immunofluorescence microscopy detected FGFR2 and FGFR4 exclusively in trophoblast cells whereas FGFR1 and FGFR3 were detected in both trophoblast cells and inner cell mass in blastocysts. Neither transcripts for FGF10 nor its receptor (FGFR2b) were temporally related to interferon τ (IFNT) transcript profile during peri- and postimplantation bovine conceptus development. A series of studies used a chemical inhibitor of FGFR kinase function (PD173074) to examine FGFR activation requirements during bovine embryo development. Exposing embryos to the inhibitor (1 µM) beginning on day 5 post-fertilization did not alter the percentage of embryos that developed into blastocysts or blastocyst cell numbers. The inhibitor did not alter the abundance of CDX2 mRNA but decreased (P<0.05) the relative abundance of IFNT mRNA in blastocysts. Exposing blastocysts to the inhibitor from days 8 to 11 post-fertilization reduced (P<0.05) the percentage of blastocysts that formed outgrowths after transfer to Matrigel-coated plates. In conclusion, each FGFR was detected in bovine embryos, and FGFR activation is needed to maximize IFNT expression and permit outgrowth formation.


Subject(s)
Embryonic Development/genetics , Fibroblast Growth Factors/pharmacology , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/physiology , Trophoblasts/drug effects , Animals , Blastocyst/metabolism , Blastocyst/physiology , Cattle/embryology , Cattle/genetics , Cattle/metabolism , Cells, Cultured , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Pregnancy , Receptors, Fibroblast Growth Factor/agonists , Receptors, Fibroblast Growth Factor/metabolism , Transcriptome , Trophoblasts/physiology
9.
Neurobiol Dis ; 48(3): 533-45, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22842016

ABSTRACT

The fibroblast growth factor receptor (FGFR) plays a vital role in the development of the nervous system regulating a multitude of cellular processes. One of the interaction partners of the FGFR is the neural cell adhesion molecule (NCAM), which is known to play an important role in neuronal development, regeneration and synaptic plasticity. Thus, simultaneous activation of FGFR- and NCAM-mediated signaling pathways may be expected to affect processes underlying neurodegenerative diseases. We here report the identification of a peptide compound, Enreptin, capable of interacting with both FGFR and NCAM. We demonstrate that this dual specificity agonist induces phosphorylation of FGFR and differentiation and survival of primary neurons in vitro, and that these effects are inhibited by abrogation of both NCAM and FGFR signaling pathways. Furthermore, Enreptin crosses the blood-brain barrier after subcutaneous administration, enhances long-term memory in normal mice and ameliorates memory deficit in mice with induced brain inflammation. Moreover, Enreptin reduces cognitive impairment and neuronal death induced by Aß25-35 in a rat model of Alzheimer's disease, and reduces the mortality rate and clinical signs of experimental autoimmune encephalomyelitis in rats. Thus, Enreptin is an attractive candidate for the treatment of neurological diseases.


Subject(s)
Memory/drug effects , Neural Cell Adhesion Molecules/agonists , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Receptors, Fibroblast Growth Factor/agonists , Animals , Behavior, Animal/drug effects , Brain Diseases/pathology , Cell Differentiation/drug effects , Cells, Cultured , Cognition Disorders/pathology , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred BALB C , Neurons/cytology , Rats , Rats, Wistar , Surface Plasmon Resonance
10.
Yao Xue Xue Bao ; 46(8): 904-9, 2011 Aug.
Article in Chinese | MEDLINE | ID: mdl-22007513

ABSTRACT

The aim of this project is to establish a fibroblast growth factor-21 (FGF-21) signaling pathway targeted cell model, for screening a class of FGF-21 receptor agonists as anti-diabetic candidates. FGF-21 requires beta klotho transmembrane proteins as co-receptor for the activation of tyrosine kinase FGF receptor (FGFR) signaling, thereby activating a series of intracellular signaling pathways and regulating gene transcription for glucose metabolism. Firstly a recombinant plasmid expressing co-receptor beta klotho and EGFP reporter genes was constructed. After introducing the recombinant plasmid into package cells, the cell culture supernatant was used to infect 3T3-L1 cells, which were then screened for stably expressing beta klotho gene. Administration of FGF-21 increased the expression of GLUT1 and stimulated GLUT1-mediated glucose uptake. This novel cell model can be conveniently used in high-throughput drug screening of FGF-21 or FGF-21 analogues.


Subject(s)
Fibroblast Growth Factors/pharmacology , Hypoglycemic Agents/metabolism , Membrane Proteins/genetics , Receptors, Fibroblast Growth Factor/agonists , 3T3-L1 Cells , Animals , Drug Evaluation, Preclinical , Fibroblast Growth Factors/metabolism , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , HEK293 Cells , Humans , Klotho Proteins , Membrane Proteins/metabolism , Mice , NIH 3T3 Cells , Plasmids , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retroviridae/genetics , Signal Transduction , Transfection
11.
J Neurochem ; 117(6): 984-94, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21480899

ABSTRACT

Neuroplastin-65 (Np65) is a brain-specific cell adhesion molecule belonging to the immunoglobulin superfamily. Homophilic trans-interaction of Np65 mediates adhesion between cells and modulates synaptic plasticity. This interaction solely occurs through the first immunoglobulin (Ig) module of Np65, but the exact binding mechanism has not yet been elucidated. In this study, we identify the homophilic binding motif of Np65 and show that a synthetic peptide modeled after this motif, termed enplastin, binds to Np65. We demonstrate that both Np65- and enplastin-induced intracellular signaling depends on fibroblast growth factor receptor, p38 mitogen-activated protein kinase, Ca(2+) /calmodulin-dependent protein kinase, and cytoplasmic Ca(2+) concentration. In addition, we show that interference with Np65 homophilic binding by enplastin has an inhibitory effect on Np65-mediated neurite outgrowth in vitro and on the initial phase of spatial learning in rats.


Subject(s)
Membrane Glycoproteins/physiology , Neuronal Plasticity , Neurons/physiology , Peptides/pharmacology , Animals , Binding Sites , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Cerebellum/cytology , Cerebellum/embryology , Cerebellum/metabolism , Enzyme Activation , Hippocampus/cytology , Hippocampus/embryology , Hippocampus/metabolism , Maze Learning/drug effects , Membrane Glycoproteins/genetics , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Mimicry , Neurites/drug effects , Neurites/physiology , Neuronal Plasticity/drug effects , Neurons/cytology , Neurons/drug effects , Peptides/genetics , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/physiology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Fibroblast Growth Factor/agonists , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
12.
Int J Mol Sci ; 11(6): 2291-305, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20640153

ABSTRACT

Fibroblast growth factor receptors (FGFRs) interact with their cognate ligands, FGFs, and with a number of cell adhesion molecules (CAMs), such as the neural cell adhesion molecule (NCAM), mediating a wide range of events during the development and maintenance of the nervous system. Determination of protein structure, in silico modeling and biological studies have recently resulted in the identification of FGFR binding peptides derived from various FGFs and NCAM mimicking the effects of these molecules with regard to their neuritogenic and neuroprotective properties. This review focuses on recently developed functional peptide agonists of FGFR with possible therapeutic potential.


Subject(s)
Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Receptors, Fibroblast Growth Factor/agonists , Animals , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Humans , Ligands , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/metabolism , Neuroprotective Agents/chemistry , Peptide Fragments/chemistry , Protein Binding , Receptors, Fibroblast Growth Factor/chemistry , Receptors, Fibroblast Growth Factor/metabolism
13.
Neurosci Res ; 68(1): 35-43, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20562017

ABSTRACT

Hexafins are recently identified low-molecular-weight peptide agonists of the fibroblast growth factor receptor (FGFR), derived from the beta6-beta7 loop region of various FGFs. Synthetic hexafin peptides have been shown to bind to and induce tyrosine phosphorylation of FGFR1, stimulate neurite outgrowth, and promote neuronal survival in vitro. Thus, the pronounced biological activities of hexafins in vitro make them attractive compounds for pharmacological studies in vivo. The present study investigated the effects of subcutaneous administration of hexafin1 and hexafin2 (peptides derived from FGF1 and FGF2, respectively) on social memory, exploratory activity, and anxiety-like behavior in adult rats. Treatment with hexafin1 and hexafin2 resulted in prolonged retention of social memory. Furthermore, rats treated with hexafin2 exhibited decreased anxiety-like behavior in the elevated plus maze. Employing an R6/2 mouse model of Huntington's disease (HD), we found that although hexafin2 did not affect the progression of motor symptoms, it alleviated deficits in activity related to social behavior, including sociability and social novelty. Thus, hexafin2 may have therapeutic potential for the treatment of HD.


Subject(s)
Behavior, Animal/drug effects , Fibroblast Growth Factors/agonists , Peptides/agonists , Peptides/pharmacology , Receptors, Fibroblast Growth Factor/agonists , Animals , Behavior, Animal/physiology , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Fibroblast Growth Factor 1/chemical synthesis , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor 2/chemical synthesis , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factors/physiology , Humans , Injections, Subcutaneous/methods , Male , Mice , Mice, Transgenic , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Peptides/chemical synthesis , Rats , Rats, Sprague-Dawley , Receptors, Fibroblast Growth Factor/physiology
14.
Neural Dev ; 5: 13, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20459606

ABSTRACT

BACKGROUND: Mouse definitive neural stem cells (NSCs) are derived from a population of LIF-responsive primitive neural stem cells (pNSCs) within the neurectoderm, yet details on the early signaling and transcriptional mechanisms that control this lineage transition are lacking. Here we tested whether FGF and Wnt signaling pathways can regulate Zfhx1b expression to control early neural stem cell development. RESULTS: By microinjecting FGF8b into the pro-amniotic cavity ex vivo at 7.0 days post-coitum (dpc) and culturing whole embryos, we demonstrate that neurectoderm-specific gene expression (for example, Sox2, Nestin, Zfhx1b) is increased, whereas Wnt3a represses neurectoderm gene expression. To determine whether FGF signaling also mediates the lineage transition from a pNSC to a NSC, 7.0-dpc embryos were microinjected with either FGF8b or inhibitors of the FGF receptor-MAP kinase signaling pathway ex vivo, cultured as whole embryos to approximately 8.5 dpc and assayed for clonal NSC colony formation. We show that pre-activation of FGF signaling in the anterior neurectoderm causes an increase in the number of colony forming NSCs derived later from the anterior neural plate, whereas inhibition of FGF signaling significantly reduces the number of NSC colonies. Interestingly, inhibition of FGF signaling causes the persistence of LIF-responsive pNSCs within the anterior neural plate and over-expression of Zfhx1b in these cells is sufficient to rescue the transition from a LIF-responsive pNSC to an FGF-responsive NSC. CONCLUSION: Our data suggest that definitive NSC fate specification in the mouse neurectoderm is facilitated by FGF activation of Zfhx1b.


Subject(s)
Ectoderm/embryology , Fibroblast Growth Factors/genetics , Homeodomain Proteins/genetics , Nervous System/embryology , Neurogenesis/genetics , Repressor Proteins/genetics , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Proliferation/drug effects , Cells, Cultured , Colony-Forming Units Assay , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/pharmacology , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Neural Tube/embryology , Neurogenesis/drug effects , Neurons/cytology , Neurons/metabolism , Receptors, Fibroblast Growth Factor/agonists , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/drug effects , Wnt Proteins/genetics , Wnt Proteins/pharmacology , Wnt3 Protein , Wnt3A Protein , Zinc Finger E-box Binding Homeobox 2
15.
J Neurosci Res ; 88(9): 1882-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20175207

ABSTRACT

Activation of fibroblast growth factor (FGF) receptors (FGFRs) both by FGFs and by the neural cell adhesion molecule (NCAM) is crucial in the development and function of the nervous system. We found that FGFR substrate 2alpha (FRS2alpha), Src homologous and collagen A (ShcA), and phospholipase-Cgamma (PLCgamma) were all required for neurite outgrowth from cerebellar granule neurons (CGNs) induced by FGF1 and FGL (an NCAM-derived peptide agonist of FGFR1). Like FGF1, FGL induced tyrosine phosphorylation of FGFR1, FRS2alpha, ShcA, and PLCgamma in a time- and dose-dependent manner. However, the activation of FRS2alpha by FGL was significantly lower than the activation by FGF1, indicating a differential signaling profile induced by NCAM compared with the cognate growth factor.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cerebellum/physiology , Fibroblast Growth Factor 1/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurons/physiology , Animals , Cell Enlargement , Cells, Cultured , Models, Neurological , Neurites/physiology , Phospholipase C gamma/metabolism , Phosphorylation , Rats , Rats, Wistar , Receptors, Fibroblast Growth Factor/agonists , Receptors, Fibroblast Growth Factor/metabolism , Shc Signaling Adaptor Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1 , Time Factors , Tyrosine/metabolism
16.
Dev Neurobiol ; 69(13): 837-54, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19634127

ABSTRACT

Fibroblast growth factor receptor (FGFR) signaling is pivotal in the regulation of neurogenesis, neuronal differentiation and survival, and synaptic plasticity both during development and in adulthood. In order to develop low molecular weight agonists of FGFR, seven peptides, termed hexafins, corresponding to the beta6-beta7 loop region of the FGF 1, 2, 3, 8, 9, 10, and 17, were synthesized. This region shares a homologous amino acid sequence with the FG-loop region of the second fibronectin Type III module of the neural cell adhesion molecule (NCAM) that binds to the FGFR. Hexafins were shown by surface plasmon resonance to bind to FGFR1-IIIc-Ig2-3 and FGFR2-IIIb-Ig2-3. The heparin analog sucrose octasulfate inhibited hexafin binding to FGFR1-IIIc-Ig2-3 indicating overlapping binding sites. Hexafin-binding to FGFR1-IIIc resulted in receptor phosphorylation, but inhibited FGF1-induced FGFR1 phosphorylation, indicating that hexafins act as partial agonists. Hexafin2, 3, 8, 10, and 17 (but not 1 or 9) induced neurite outgrowth from cerebellar granule neurons (CGNs), an effect that was abolished by two inhibitors of FGFR, SU5402 and inositol hexaphosphate (IP6) and a diacylglycerol lipase inhibitor, RHC-80267. The neuritogenic effects of selected hexafins could also be inhibited by FGF1 which by itself did not induce neurite outgrowth. Moreover, hexafin1, 3, 9, 10, and 17 (but not 2 or 8) promoted survival of CGNs induced to undergo apoptosis. Thus, selected hexafins induced neuronal differentiation and survival, making them promising pharmacological tools for the study of functional FGFR regulation in development of the nervous system.


Subject(s)
Cell Survival/drug effects , Fibroblast Growth Factors/metabolism , Neurons/cytology , Receptors, Fibroblast Growth Factor/metabolism , Analysis of Variance , Animals , Binding Sites , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Neural Cell Adhesion Molecules/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Phytic Acid/pharmacology , Pyrroles/pharmacology , Rats , Rats, Wistar , Receptors, Fibroblast Growth Factor/agonists , Video Recording
17.
J Neurosci Res ; 87(8): 1806-12, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19185025

ABSTRACT

The neural cell adhesion molecule L1 plays an important role in axon growth, neuronal survival, and synaptic plasticity. We recently demonstrated that the L1 fibronectin type III (FN3) modules interact directly with the fibroblast growth factor (FGF) receptor (FGFR). Sequence alignment of individual L1 FN3 modules with various FGFs suggested that four sequence motifs located in the third and fifth L1 FN3 modules might be involved in interactions with FGFR. The present study found that corresponding synthetic peptides, termed elcamins 1, 2, 3, and 4, bind and activate FGFR in the absence of FGF1. Conversely, in the presence of FGF1, elcamins inhibited receptor phosphorylation, indicating that the peptides are FGFR partial agonists. Elcamins 1, 3, and 4 dose dependently induced neurite outgrowth in cultured primary cerebellar neurons. The neuritogenic effect of elcamins was dependent on FGFR activation, insofar as the effect was abolished by the receptor inhibition. Thus, the identified peptides act as L1 mimetics with regard to activation of FGFR and induction of neurite outgrowth.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/embryology , Central Nervous System/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurogenesis/physiology , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Motifs/physiology , Amino Acid Sequence/physiology , Animals , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Ligands , Neural Cell Adhesion Molecule L1/agonists , Neural Cell Adhesion Molecule L1/chemistry , Neurites/drug effects , Neurites/metabolism , Neurites/ultrastructure , Peptides/metabolism , Peptides/pharmacology , Phosphorylation/drug effects , Protein Binding/physiology , Rats , Receptors, Fibroblast Growth Factor/agonists , Up-Regulation/drug effects , Up-Regulation/physiology
18.
Eur J Neurosci ; 28(8): 1618-28, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18973581

ABSTRACT

The neural cell adhesion molecule (NCAM) plays a pivotal role in brain plasticity. Brain plasticity itself has a crucial role in the development of depression. The aim of this study was to analyze whether NCAM-deficient (NCAM(-/-)) mice exhibit depression-like behaviour and whether a peptide termed FGL, derived from the NCAM binding site for the fibroblast growth factor (FGF) receptor, is able to reverse the depression-like signs in NCAM(-/-) mice. Our study showed that NCAM(-/-) mice demonstrated increased freezing time in the tail-suspension test and reduced preference for sucrose consumption in the sucrose preference test, reduced adult neurogenesis in the dentate gyrus and reduced levels of the phosphorylated cAMP response element-binding protein (pCREB) in the hippocampus. FGL administered acutely or repeatedly reduced depression-like behaviour in NCAM(-/-) mice without having an effect on their wild-type littermates. Repeated administration of FGL enhanced survival of the newly born neurons in NCAM(-/-) mice and increased the levels of pCREB in both NCAM(+/+) and NCAM(-/-) mice. In conclusion, our data demonstrate that NCAM deficiency in mice results in a depression-like phenotype which can be reversed by the acute or repeated administration of FGL. The results also suggest a role of the deficit in NCAM signalling through the FGF receptor in depression.


Subject(s)
Depressive Disorder/drug therapy , Depressive Disorder/genetics , Neural Cell Adhesion Molecules/agonists , Neural Cell Adhesion Molecules/genetics , Receptors, Fibroblast Growth Factor/agonists , Animals , Atrophy/drug therapy , Atrophy/physiopathology , Atrophy/prevention & control , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Cell Survival/drug effects , Cell Survival/genetics , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Depressive Disorder/physiopathology , Disease Models, Animal , Fibroblast Growth Factors/agonists , Fibroblast Growth Factors/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Mice , Mice, Knockout , Neural Cell Adhesion Molecules/pharmacology , Neural Cell Adhesion Molecules/therapeutic use , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Neurons/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/physiology
19.
Biochim Biophys Acta ; 1780(12): 1432-40, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18760333

ABSTRACT

Structural instability of wild-type fibroblast growth factor (FGF)-1 and its dependence on exogenous heparin for optimal activity diminishes its potential utility as a therapeutic agent. Here we evaluated FGFC, an FGF1:FGF2 chimeric protein, for its receptor affinity, absolute heparin-dependence, stability and potential clinical applicability. Using BaF3 transfectants overexpressing each FGF receptor (FGFR) subtype, we found that, like FGF1, FGFC activates all of the FGFR subtypes (i.e., FGFR1c, FGFR1b, FGFR2c, FGFR2b, FGFR3c, FGFR3b and FGFR4) in the presence of heparin. Moreover, FGFC activates FGFRs even in the absence of heparin. FGFC stimulated keratinocytes proliferation much more strongly than FGF2, as would be expected from its ability to activate FGFR2b. FGFC showed greater structural stability, biological activity and resistance to trypsinization, and less loss in solution than FGF1 or FGF2. When FGFC was intraperitoneally administered to BALB/c mice prior to whole body gamma-irradiation, survival of small intestine crypts was significantly enhanced, as compared to control mice. These results suggest that FGFC could be useful in a variety of clinical applications, including promotion of wound healing and protection against radiation-induced damage.


Subject(s)
Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 2/genetics , Radiation-Protective Agents/pharmacology , Receptors, Fibroblast Growth Factor/agonists , Recombinant Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , Cell Line , Cell Proliferation/drug effects , Fibroblast Growth Factor 1/chemistry , Fibroblast Growth Factor 2/chemistry , Gamma Rays , Heparin/pharmacology , Intestine, Small/drug effects , Intestine, Small/pathology , Intestine, Small/radiation effects , Keratinocytes/cytology , Keratinocytes/drug effects , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Folding , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Solutions , Trypsin/metabolism , Whole-Body Irradiation
20.
Protein Sci ; 17(10): 1698-705, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18593816

ABSTRACT

The fibroblast growth factor receptor (FGFR) can be activated through direct interaction with the neural cell adhesion molecule (NCAM). The extracellular part of the FGFR consists of three immunoglobulin-like (Ig) modules, and that of the NCAM consists of five Ig and two fibronectin type III (F3) modules. NCAM-FGFR interactions are mediated by the third FGFR Ig module and the second NCAM F3 module. Using surface plasmon resonance and nuclear magnetic resonance analyses, the present study demonstrates that the second Ig module of FGFR also is involved in binding to the NCAM. The second Ig module residues involved in binding were identified and shown to be localized on the "opposite sides" of the module, indicating that when NCAMs are clustered (e.g., due to homophilic binding), high-affinity FGFR binding sites may be formed by the neighboring NCAMs.


Subject(s)
Models, Molecular , Neural Cell Adhesion Molecules/chemistry , Receptors, Fibroblast Growth Factor/agonists , Receptors, Fibroblast Growth Factor/chemistry , Animals , Binding Sites , Mice , Neural Cell Adhesion Molecules/metabolism , Protein Binding , Protein Conformation , Protein Interaction Mapping , Rats , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...