Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 192: 114678, 2021 10.
Article in English | MEDLINE | ID: mdl-34265279

ABSTRACT

Fibroblast growth factor 21 (FGF21) is mainly regulated by peroxisome proliferator-activated receptor α (PPARα) in liver. The PPARα-FGF21 axis protects against alcohol-related liver disease (ALD). FGF21 exerts its effect via FGF receptor 1 (FGFR1). However, liver specific FGFR1 abrogation had no effect on ALD. Adipose tissues highly express FGFR1. When adipocyte specific FGFR1 knockout (fgfr1adipoQ-cre) mice and corresponding normal control (fgfr1fl/fl) mice were fed with Lieber-DeCarli ethanol liquid diet for 3 weeks, liver triglyceride (TG) accumulation was increased in the fgfr1fl/fl mice to a greater extent than in the fgfr1adipoQ-cre mice. When PPARα agonist WY-14,643 was added in the liquid ethanol diet at 10 mg/L, the ethanol-induced liver TG accumulation was blunted in the fgfr1fl/fl mice but not in the fgfr1adipoQ-cre mice. There was no significant difference in WY-14,643-induced fatty acid oxidation, ethanol metabolism, and oxidative stress between the fgfr1fl/fl and fgfr1adipoQ-cre mice. Interestingly, adipose atrophy was induced by WY-14,643 in the fgfr1adipoQ-cre mice but not in the fgfr1fl/fl mice. Serum free fatty acid was also decreased by WY-14,643 in the fgfr1adipoQ-cre mice but not in the fgfr1fl/fl mice. These results suggest that WY-14,643 inhibits alcoholic fatty liver and regulates adipose tissue mass and fat mobilization from adipose tissues to liver in an adipocyte FGFR1-dependent manner.


Subject(s)
Adipose Tissue/drug effects , Ethanol/toxicity , Fatty Liver, Alcoholic/prevention & control , PPAR alpha/agonists , Pyrimidines/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Adipose Tissue/metabolism , Animals , Atrophy/chemically induced , Atrophy/metabolism , Ethanol/administration & dosage , Fatty Liver, Alcoholic/metabolism , Female , Mice , Mice, Knockout , PPAR alpha/metabolism , Peroxisome Proliferators/therapeutic use , Peroxisome Proliferators/toxicity , Pyrimidines/toxicity , Receptor, Fibroblast Growth Factor, Type 1/genetics
2.
Nature ; 590(7846): 504-508, 2021 02.
Article in English | MEDLINE | ID: mdl-33536620

ABSTRACT

Amplification of chromosomal region 8p11-12 is a common genetic alteration that has been implicated in the aetiology of lung squamous cell carcinoma (LUSC)1-3. The FGFR1 gene is the main candidate driver of tumorigenesis within this region4. However, clinical trials evaluating FGFR1 inhibition as a targeted therapy have been unsuccessful5. Here we identify the histone H3 lysine 36 (H3K36) methyltransferase NSD3, the gene for which is located in the 8p11-12 amplicon, as a key regulator of LUSC tumorigenesis. In contrast to other 8p11-12 candidate LUSC drivers, increased expression of NSD3 correlated strongly with its gene amplification. Ablation of NSD3, but not of FGFR1, attenuated tumour growth and extended survival in a mouse model of LUSC. We identify an LUSC-associated variant NSD3(T1232A) that shows increased catalytic activity for dimethylation of H3K36 (H3K36me2) in vitro and in vivo. Structural dynamic analyses revealed that the T1232A substitution elicited localized mobility changes throughout the catalytic domain of NSD3 to relieve auto-inhibition and to increase accessibility of the H3 substrate. Expression of NSD3(T1232A) in vivo accelerated tumorigenesis and decreased overall survival in mouse models of LUSC. Pathological generation of H3K36me2 by NSD3(T1232A) reprograms the chromatin landscape to promote oncogenic gene expression signatures. Furthermore, NSD3, in a manner dependent on its catalytic activity, promoted transformation in human tracheobronchial cells and growth of xenografted human LUSC cell lines with amplification of 8p11-12. Depletion of NSD3 in patient-derived xenografts from primary LUSCs containing NSD3 amplification or the NSD3(T1232A)-encoding variant attenuated neoplastic growth in mice. Finally, NSD3-regulated LUSC-derived xenografts were hypersensitive to bromodomain inhibition. Thus, our work identifies NSD3 as a principal 8p11-12 amplicon-associated oncogenic driver in LUSC, and suggests that NSD3-dependency renders LUSC therapeutically vulnerable to bromodomain inhibition.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Histones/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Nuclear Proteins/metabolism , Animals , Biocatalysis , Carcinogenesis/genetics , Carcinoma, Squamous Cell/genetics , Female , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Humans , Lung Neoplasms/genetics , Male , Methylation , Mice , Models, Molecular , Mutation , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Xenograft Model Antitumor Assays
3.
Hypertension ; 76(6): 1935-1944, 2020 12.
Article in English | MEDLINE | ID: mdl-33131311

ABSTRACT

Endothelial-to-mesenchymal transition (EndMT) has been shown to contribute to organ fibrogenesis. We have reported that N-acetyl-seryl-aspartyl- lysyl-proline (AcSDKP) restored levels of diabetes mellitus-suppressed FGFR1 (fibroblast growth factor receptor 1), the endothelial receptor essential for combating EndMT. However, the molecular regulation and biological/pathological significance of the AcSDKP-FGFR1 relationship has not been elucidated yet. Here, we demonstrated that endothelial FGFR1 deficiency led to AcSDKP-resistant EndMT and severe fibrosis associated with EndMT-stimulated fibrogenic programming in neighboring cells. Diabetes mellitus induced severe kidney fibrosis in endothelial FGFR1-deficient mice (FGFR1fl/fl; VE-cadherin-Cre: FGFR1EKO) but not in control mice (FGFR1fl/fl); AcSDKP completely or partially suppressed kidney fibrosis in control or FGFR1EKO mice. Severe fibrosis was also induced in hearts of diabetic FGFR1EKO mice; however, AcSDKP had no effect on heart fibrosis in FGFR1EKO mice. AcSDKP also had no effect on EndMT in either kidney or heart but partially suppressed epithelial-to-mesenchymal transition in kidneys of diabetic FGFR1EKO mice. The medium from FGFR1-deficient endothelial cells stimulated TGFß (transforming growth factor ß)/Smad-dependent epithelial-to-mesenchymal transition in cultured human proximal tubule epithelial cell line, AcSDKP inhibited such epithelial-to-mesenchymal transition. These data demonstrated that endothelial FGFR1 is essential as an antifibrotic core molecule as the target of AcSDKP.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endothelium/metabolism , Kidney/metabolism , Myocardium/metabolism , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Animals , Cell Line , Diabetes Mellitus, Experimental/pathology , Endothelium/cytology , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Fibrosis , Humans , Kidney/pathology , Kidney Tubules, Proximal/cytology , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Oligopeptides/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/genetics
4.
J Bone Miner Res ; 34(9): 1660-1675, 2019 09.
Article in English | MEDLINE | ID: mdl-31206783

ABSTRACT

Fibroblast growth factor (FGF) signaling pathways have well-established roles in skeletal development, with essential functions in both chondrogenesis and osteogenesis. In mice, previous conditional knockout studies suggested distinct roles for FGF receptor 1 (FGFR1) signaling at different stages of osteogenesis and a role for FGFR2 in osteoblast maturation. However, the potential for redundancy among FGFRs and the mechanisms and consequences of stage-specific osteoblast lineage regulation were not addressed. Here, we conditionally inactivate Fgfr1 and Fgfr2 in mature osteoblasts with an Osteocalcin (OC)-Cre or Dentin matrix protein 1 (Dmp1)-CreER driver. We find that young mice lacking both receptors or only FGFR1 are phenotypically normal. However, between 6 and 12 weeks of age, OC-Cre Fgfr1/Fgfr2 double- and Fgfr1 single-conditional knockout mice develop a high bone mass phenotype with increased periosteal apposition, increased and disorganized endocortical bone with increased porosity, and biomechanical properties that reflect increased bone mass but impaired material properties. Histopathological and gene expression analyses show that this phenotype is preceded by a striking loss of osteocytes and accompanied by activation of the Wnt/ß-catenin signaling pathway. These data identify a role for FGFR1 signaling in mature osteoblasts/osteocytes that is directly or indirectly required for osteocyte survival and regulation of bone mass during postnatal bone growth. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Bone Development , Osteoblasts/metabolism , Osteocytes/pathology , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Alleles , Animals , Biomechanical Phenomena , Bone Remodeling , Cell Death , Cell Survival , Cortical Bone/pathology , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Osteoblasts/pathology , Osteocytes/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Wnt Signaling Pathway
5.
Nature ; 545(7653): 224-228, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28467822

ABSTRACT

Blood and lymphatic vasculatures are intimately involved in tissue oxygenation and fluid homeostasis maintenance. Assembly of these vascular networks involves sprouting, migration and proliferation of endothelial cells. Recent studies have suggested that changes in cellular metabolism are important to these processes. Although much is known about vascular endothelial growth factor (VEGF)-dependent regulation of vascular development and metabolism, little is understood about the role of fibroblast growth factors (FGFs) in this context. Here we identify FGF receptor (FGFR) signalling as a critical regulator of vascular development. This is achieved by FGF-dependent control of c-MYC (MYC) expression that, in turn, regulates expression of the glycolytic enzyme hexokinase 2 (HK2). A decrease in HK2 levels in the absence of FGF signalling inputs results in decreased glycolysis, leading to impaired endothelial cell proliferation and migration. Pan-endothelial- and lymphatic-specific Hk2 knockouts phenocopy blood and/or lymphatic vascular defects seen in Fgfr1/Fgfr3 double mutant mice, while HK2 overexpression partly rescues the defects caused by suppression of FGF signalling. Thus, FGF-dependent regulation of endothelial glycolysis is a pivotal process in developmental and adult vascular growth and development.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibroblast Growth Factors/metabolism , Glycolysis , Neovascularization, Physiologic , Signal Transduction , Animals , Cell Movement , Cell Proliferation , Female , Hexokinase/metabolism , Lymphangiogenesis , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-myc/metabolism , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 3/deficiency , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism
6.
Am J Physiol Heart Circ Physiol ; 310(5): H559-71, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26747503

ABSTRACT

Fibroblast growth factor (FGF) signaling is cardioprotective in various models of myocardial infarction. FGF receptors (FGFRs) are expressed in multiple cell types in the adult heart, but the cell type-specific FGFR signaling that mediates different cardioprotective endpoints is not known. To determine the requirement for FGFR signaling in endothelium in cardiac ischemia-reperfusion injury, we conditionally inactivated the Fgfr1 and Fgfr2 genes in endothelial cells with Tie2-Cre (Tie2-Cre, Fgfr1(f/f), Fgfr2(f/f) DCKO mice). Tie2-Cre, Fgfr1(f/f), Fgfr2(f/f) DCKO mice had normal baseline cardiac morphometry, function, and vessel density. When subjected to closed-chest, regional cardiac ischemia-reperfusion injury, Tie2-Cre, Fgfr1(f/f), Fgfr2(f/f) DCKO mice showed a significantly increased hypokinetic area at 7 days, but not 1 day, after reperfusion. Tie2-Cre, Fgfr1(f/f), Fgfr2(f/f) DCKO mice also showed significantly worsened cardiac function compared with controls at 7 days but not 1 day after reperfusion. Pathophysiological analysis showed significantly decreased vessel density, increased endothelial cell apoptosis, and worsened tissue hypoxia in the peri-infarct area at 7 days following reperfusion. Notably, Tie2-Cre, Fgfr1(f/f), Fgfr2(f/f) DCKO mice showed no impairment in the cardiac hypertrophic response. These data demonstrate an essential role for FGFR1 and FGFR2 in endothelial cells for cardiac functional recovery and vascular remodeling following in vivo cardiac ischemia-reperfusion injury, without affecting the cardiac hypertrophic response. This study suggests the potential for therapeutic benefit from activation of endothelial FGFR pathways following ischemic injury to the heart.


Subject(s)
Endothelial Cells/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Vascular Remodeling , Animals , Apoptosis , Capillaries/metabolism , Capillaries/pathology , Capillaries/physiopathology , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Disease Models, Animal , Endothelial Cells/pathology , Female , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Neovascularization, Physiologic , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptor, Fibroblast Growth Factor, Type 2/genetics , Recovery of Function , Signal Transduction , Stroke Volume , Time Factors , Ventricular Function, Left
7.
Oncogene ; 35(22): 2852-61, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26387545

ABSTRACT

Osteosarcoma is the most common primary malignancy of the skeleton and is prevalent in children and adolescents. Survival rates are poor and have remained stagnant owing to chemoresistance and the high propensity to form lung metastases. In this study, we used in vivo transgenic models of c-fos oncogene-induced osteosarcoma and chondrosarcoma in addition to c-Fos-inducible systems in vitro to investigate downstream signalling pathways that regulate osteosarcoma growth and metastasis. Fgfr1 (fibroblast growth factor receptor 1) was identified as a novel c-Fos/activator protein-1(AP-1)-regulated gene. Induction of c-Fos in vitro in osteoblasts and chondroblasts caused an increase in Fgfr1 RNA and FGFR1 protein expression levels that resulted in increased and sustained activation of mitogen-activated protein kinases (MAPKs), morphological transformation and increased anchorage-independent growth in response to FGF2 ligand treatment. High levels of FGFR1 protein and activated pFRS2α signalling were observed in murine and human osteosarcomas. Pharmacological inhibition of FGFR1 signalling blocked MAPK activation and colony growth of osteosarcoma cells in vitro. Orthotopic injection in vivo of FGFR1-silenced osteosarcoma cells caused a marked twofold to fivefold decrease in spontaneous lung metastases. Similarly, inhibition of FGFR signalling in vivo with the small-molecule inhibitor AZD4547 markedly reduced the number and size of metastatic nodules. Thus deregulated FGFR signalling has an important role in osteoblast transformation and osteosarcoma formation and regulates the development of lung metastases. Our findings support the development of anti-FGFR inhibitors as potential antimetastatic therapy.


Subject(s)
Lung Neoplasms/secondary , Osteosarcoma/pathology , Proto-Oncogene Proteins c-fos/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Transcription Factor AP-1/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chondrocytes/drug effects , Chondrocytes/pathology , Chondrosarcoma/genetics , Chondrosarcoma/pathology , Colon/drug effects , Colon/pathology , Enzyme Activation/drug effects , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Oncogenes/genetics , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteosarcoma/genetics , Proto-Oncogene Proteins c-fos/genetics , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects , Transcriptional Activation
8.
Eur J Immunol ; 45(9): 2517-28, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26081170

ABSTRACT

Chronic skin inflammation resulting from a defective epidermal barrier is a hallmark of atopic dermatitis (AD). We previously demonstrated that mice lacking FGF receptors 1 and 2 in keratinocytes (K5-R1/R2 mice) develop an AD-like chronic dermatitis as a result of an impaired epidermal barrier. Here, we show that γδ T cells, which rapidly respond to various insults, accumulate in the epidermis of K5-R1/R2 mice before the development of histological abnormalities. Their number and activation further increase as the phenotype progresses, most likely as a consequence of increased expression of Il-2 and Il-7 and the stress-induced proteins Rae-1, H60c, Mult1, PlexinB2, and Skint1. To determine the role of γδ T cells in the skin phenotype, we generated quadruple mutant K5-R1/-R2 mice lacking γδ T cells. Surprisingly, loss of γδ T cells did not or only marginally affect keratinocyte proliferation, epidermal thickness, epidermal barrier function, and accumulation and activation of different immune cells in the skin of K5-R1/R2 mice, possibly due to partial compensation by αß T cells. These results demonstrate that γδ T cells do not contribute to the development or maintenance of chronic inflammation in response to a defect in the epidermal barrier.


Subject(s)
Dermatitis/immunology , Keratinocytes/immunology , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptors, Antigen, T-Cell, gamma-delta/deficiency , T-Lymphocyte Subsets/immunology , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Proliferation , Chronic Disease , Dermatitis/genetics , Dermatitis/pathology , Disease Models, Animal , Epidermis/immunology , Epidermis/pathology , Gene Expression Regulation , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Immunoglobulins/genetics , Immunoglobulins/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-7/genetics , Interleukin-7/immunology , Keratinocytes/pathology , Lymphocyte Activation , Lymphocyte Depletion , Membrane Proteins , Mice , Mice, Transgenic , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/immunology , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/immunology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/immunology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Signal Transduction , T-Lymphocyte Subsets/pathology
9.
Dev Dyn ; 244(6): 759-73, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25820239

ABSTRACT

BACKGROUND: The developmental processes of the genital tubercle (GT), the anlage of the external genitalia, possess several developmental aspects, including GT outgrowth, urethral tube formation, and epithelial differentiation of the urethra. The GT comprises the mesenchyme derived from the lateral mesoderm, ectodermal epithelium, and endodermal epithelium (embryonic urethral epithelium). The three tissue layers develop the GT coordinately. RESULTS: Around the initial stage of GT outgrowth (E11.5), FGF signaling was detected in the mesenchyme of the GT. FGF signaling was detected in the three tissue layers of the GT around the early stage of urethral formation (E13.5). Subsequently, FGF signaling was predominantly detected in the urethral epithelium (E14.5). Tissue-specific roles of FGF signaling in GT development were revealed by conditional Fgfr gene knockout approaches. Mesenchymal FGF signaling in the early-stage GT is required for its outgrowth. Ectodermal FGF signaling in the GT is required for the differentiation of the ectoderm and urethral epithelium at their junction to form the proper urethral tube. Endodermal FGF signaling in the GT is required for the stratification and cell adhesive characteristics of the urethral epithelium. CONCLUSIONS: The current study suggests that spatiotemporally regulated FGF signaling plays tissue-specific roles in multiple processes of external genitalia development.


Subject(s)
Fibroblast Growth Factors/physiology , Genitalia, Female/embryology , Genitalia, Male/embryology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 2/physiology , Animals , Bone Morphogenetic Proteins/physiology , Cell Adhesion , Cell Survival , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Genitalia, Female/physiology , Genitalia, Male/physiology , Germ Layers/cytology , Germ Layers/metabolism , Hedgehog Proteins/physiology , Male , Mesoderm/physiology , Mice , Mice, Knockout , Organ Specificity , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptor, Fibroblast Growth Factor, Type 2/genetics , Recombinant Fusion Proteins/metabolism , Urethra/embryology , Urethra/physiology , Wnt Proteins/physiology , Wnt Signaling Pathway
10.
Sci Signal ; 7(344): ra90, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25249657

ABSTRACT

Abnormal vascular homeostasis can lead to increased proliferation of smooth muscle cells and deposition of extracellular matrix, resulting in neointima formation, which contributes to vascular lumen narrowing, a pathology that underlies diseases including transplant vasculopathy, the recurrence of stenosis, and atherosclerosis. Growth of neointima is in part due to endothelial-to-mesenchymal transition (EndMT), a transforming growth factor-ß (TGFß)-driven process, which leads to increased numbers of smooth muscle cells and fibroblasts and deposition of extracellular matrix. We reported that endothelial cell-specific knockout of fibroblast growth factor receptor 1 (FGFR1) led to activation of TGFß signaling and development of EndMT in vitro and in vivo. Furthermore, EndMT in human diseased vasculature correlated with decreased abundance of FGFR1. These findings identify FGFR1 as the key regulator of TGFß signaling and EndMT development.


Subject(s)
Endothelium, Vascular/physiology , Neointima/physiopathology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Animals , Cell Transdifferentiation , Coronary Vessels/metabolism , Coronary Vessels/pathology , Coronary Vessels/transplantation , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Graft Rejection/pathology , Heart Transplantation , Heterografts , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Ischemia/metabolism , Mesoderm/cytology , Mice , Mice, Mutant Strains , MicroRNAs/metabolism , Muscle, Smooth, Vascular/pathology , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Smad2 Protein/metabolism , Transplantation Chimera
11.
Behav Brain Res ; 264: 74-81, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24512770

ABSTRACT

Serotonergic neurons in the dorsal raphe nucleus (DR) are organized in anatomically distinct subregions that form connections with specific brain structures to modulate diverse behaviors, including anxiety-like behavior. It is unclear if the functional heterogeneity of these neurons is coupled to their developmental heterogeneity, and if abnormal development of specific DR serotonergic subregions can permanently impact anxiety circuits and behavior. The goal of this study was to examine if deficiencies in different components of fibroblast growth factor (Fgf) signaling could preferentially impact the development of specific populations of DR serotonergic neurons to alter anxiety-like behavior in adulthood. Wild-type and heterozygous male mice globally hypomorphic for Fgf8, Fgfr1, or both (Fgfr1/Fgf8) were tested in an anxiety-related behavioral battery. Both Fgf8- and Fgfr1/Fgf8-deficient mice display increased anxiety-like behavior as measured in the elevated plus-maze and the open-field tests. Immunohistochemical staining of a serotonergic marker, tryptophan hydroxylase (Tph), revealed reductions in specific populations of serotonergic neurons in the ventral, interfascicular, and ventrolateral/ventrolateral periaqueductal gray subregions of the DR in all Fgf-deficient mice, suggesting a neuroanatomical basis for increased anxiety-like behavior. Overall, this study suggests Fgf signaling selectively modulates the development of different serotonergic neuron subpopulations. Further, it suggests anxiety-like behavior may stem from developmental disruption of these neurons, and individuals with inactivating mutations in Fgf signaling genes may be predisposed to anxiety disorders.


Subject(s)
Adaptation, Psychological/physiology , Anxiety/genetics , Brain/metabolism , Fibroblast Growth Factor 8/deficiency , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Serotonin/metabolism , Analysis of Variance , Animals , Anxiety/physiopathology , Brain/cytology , Exploratory Behavior/physiology , Fibroblast Growth Factor 8/genetics , Gene Expression Regulation/genetics , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Muscle Strength/genetics , Mutation/genetics , Neurons/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Tryptophan Hydroxylase/metabolism
13.
Cell Res ; 23(2): 254-73, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23147794

ABSTRACT

The optic fissure (OF) is a transient opening on the ventral side of the developing vertebrate eye that closes before nearly all retinal progenitor cell differentiation has occurred. Failure to close the OF results in coloboma, a congenital disease that is a major cause of childhood blindness. Although human genetic studies and animal models have linked a number of genes to coloboma, the cellular and molecular mechanisms driving the closure of the OF are still largely unclear. In this study, we used Cre-LoxP-mediated conditional removal of fibroblast growth factor (FGF) receptors, Fgfr1 and Fgfr2, from the developing optic cup (OC) to show that FGF signaling regulates the closing of the OF. Our molecular, cellular and transcriptome analyses of Fgfr1 and Fgfr2 double conditional knockout OCs suggest that FGF signaling controls the OF closure through modulation of retinal progenitor cell proliferation, fate specification and morphological changes. Furthermore, Fgfr1 and Fgfr2 double conditional mutant retinal progenitor cells fail to initiate retinal ganglion cell (RGC) genesis. Taken together, our mouse genetic studies reveal that FGF signaling is essential for OF morphogenesis and RGC development.


Subject(s)
Coloboma/metabolism , Fibroblast Growth Factors/metabolism , Retina/cytology , Signal Transduction , Animals , Coloboma/pathology , Gene Expression Profiling , Mice , Mice, Knockout , Neurogenesis , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism
14.
Brain Res ; 1460: 12-24, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22578469

ABSTRACT

Fibroblast growth factor receptor (FGFR) signaling determines the size of the cerebral cortex by regulating the amplification of radial glial stem cells, and participates in the formation of midline glial structures. We show that Fgfr1 and Fgfr2 double knockouts (FGFR DKO) generated by Cre-mediated recombination driven by the human GFAP promoter (hGFAP) have reduced cerebellar size due to reduced proliferation of radial glia and other glial precursors in late embryonic and neonatal FGFR DKO mice. The proliferation of granule cell progenitors (GCPs) in the EGL was also reduced, leading to reduced granule cell numbers. Furthermore, both inward migration of granule cells into the inner granule cell layer (IGL) and outward migration of GABA interneurons into the molecular layer (ML) were arrested, disrupting layer and lobular morphology. Purkinje neurons and their dendrites, which were not targeted by Cre-mediated recombination of Fgf receptors, were also misplaced in FGFR DKO mice, possibly as a consequence of altered Bergmann glia orientation or reduced granule cell number. Our findings indicate a dual role for FGFR signaling in cerebellar morphogenesis. The first role is to amplify the number of granule neuron precursors in the external granular layer and glial precursor cells throughout the cerebellum. The second is to establish the correct Bergmann glia morphology, which is crucial for granule cell migration. The disrupted cerebellar size and laminar architecture resulting from loss of FGFR signaling impair motor learning and coordination in FGFR DKO mice.


Subject(s)
Cerebellar Diseases/genetics , Cerebellar Diseases/pathology , Motor Skills Disorders/pathology , Motor Skills Disorders/physiopathology , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Animals , Cerebellar Diseases/physiopathology , Female , Humans , Male , Mice , Mice, Knockout , Mice, Neurologic Mutants , Motor Skills Disorders/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism
15.
Schizophr Res ; 136(1-3): 82-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22285656

ABSTRACT

Genetic and post mortem evidence has implicated the α7 neuronal nicotinic receptor (NNR) in the etiology of schizophrenia and related disorders. In schizophrenia, enhanced subcortical dopamine (DA) correlates with positive and cognitive of the disease, including impairments in sensorimotor gating. We measured the levels of extracellular DA and DA metabolites during an acoustic test session of prepulse inhibition (PPI) of the startle response, a measure of sensorimotor gating, by microdialysis and HPLC-EC in a transgenic mouse model of schizophrenia. In th-fgfr1(tk-) mice, blockade of fibroblast growth factor receptor 1 (FGFR1) signaling during development in catecholaminergic neurons results in reduced size and density of midbrain DA neurons of the substantia nigra pars compacta (SNc) and ventral tegmental area (VTA). These mice displayed reduced PPI and enhanced startle response relative to control mice as well as a potentiation of DA release in the dorsal striatum during a 30 minute PPI test session. Acute administration of a partial α7 NNR agonist TC-7020 (1.0 mg/kg) normalized PPI and startle deficits and attenuated increases of DA release during acoustic PPI testing. These results provide direct evidence of elevated striatal dopaminergic transmission with impaired sensorimotor gating that may underlie cognitive and positive symptoms and motor deficits in schizophrenia and related disorders. Also, systemic targeting of alpha7 NNRs may ameliorate these deficits by functionally suppressing striatal DA activity.


Subject(s)
Corpus Striatum/drug effects , Dopamine/metabolism , Neural Inhibition/drug effects , Schizophrenia/drug therapy , Schizophrenia/physiopathology , Sensory Gating/drug effects , 3,4-Dihydroxyphenylacetic Acid/metabolism , Acoustic Stimulation/methods , Animals , Chromatography, High Pressure Liquid , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , Homovanillic Acid/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microdialysis/methods , Neural Inhibition/genetics , Nicotinic Agonists/pharmacology , Quinuclidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Reflex, Startle/drug effects , Reflex, Startle/genetics , Schizophrenia/genetics , Sensory Gating/genetics , Thiophenes/pharmacology
16.
Circ Res ; 110(4): e29-39, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22207710

ABSTRACT

RATIONALE: Although the fibroblast growth factor (FGF) signaling axis plays important roles in heart development, the molecular mechanism by which the FGF regulates cardiogenesis is not fully understood. OBJECTIVE: To investigate the mechanism by which FGF signaling regulates cardiac progenitor cell differentiation. METHODS AND RESULTS: Using mice with tissue-specific ablation of FGF receptors and FGF receptor substrate 2α (Frs2α) in heart progenitor cells, we demonstrate that disruption of FGF signaling leads to premature differentiation of cardiac progenitor cells in mice. Using embryoid body cultures of mouse embryonic stem cells, we reveal that FGF signaling promotes mesoderm differentiation in embryonic stem cells but inhibits cardiomyocyte differentiation of the mesoderm cells at later stages. Furthermore, we also report that inhibiting FRS2α-mediated signals increases autophagy and that activating autophagy promotes myocardial differentiation and vice versa. CONCLUSIONS: The results indicate that the FGF/FRS2α-mediated signals prevent premature differentiation of heart progenitor cells through suppressing autophagy. The findings provide the first evidence that autophagy plays a role in heart progenitor differentiation.


Subject(s)
Autophagy , Cell Differentiation , Fibroblast Growth Factors/metabolism , Heart/embryology , Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction , Stem Cells/metabolism , Animals , Cell Proliferation , Cells, Cultured , Embryo Culture Techniques , Gene Expression Regulation, Developmental , Genotype , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Phenotype , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptor, Fibroblast Growth Factor, Type 2/genetics , Stem Cells/pathology , Time Factors , Tissue Culture Techniques
17.
J Neurosci ; 31(13): 5055-66, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21451043

ABSTRACT

Fibroblast growth factors (FGFs) comprise a family of developmental regulators implicated in a wide variety of neurological functions. FGF receptors 1, 2, and 3 (Fgfrs) are expressed in the embryonic forebrain, including regions overlapping with ventral sites of oligodendrocyte progenitor (OLP) generation. Although FGF signaling is known to influence the proliferation of OLPs in vitro, functions of different Fgfrs in vivo are lacking. Here, we examined single and double mutants with conditional disruption of Fgfrs, specifically in the embryonic forebrain, to investigate the effect of FGFs on the generation and proliferation of OLPs in vivo. FGF signaling, through cooperation between Fgfr1 and Fgfr2 but not Fgfr3, is required for the initial generation of OLPs in the mouse ventral forebrain, with Fgfr1 being a stronger inducer than Fgfr2. In cultures derived from embryonic mutant forebrains or from normal forebrains grown in the presence of Fgfr inhibitor, a strong attenuation of OLP generation was observed, supporting the role of FGF signaling in vivo. Contrary to in vitro findings, Fgfr1 and Fgfr2 signaling is not required for the proliferation of OLPs in vivo. Finally, failure of OLP generation in the Fgfr mutants occurred without loss of sonic hedgehog (Shh) signaling; and pharmacological inhibition of either Fgfr or hedgehog signaling in parallel cultures strongly inhibited OLP generation, suggesting that Fgfrs cooperate with Shh to generate OLPs. Overall, our results reveal for the first time an essential role of FGF signaling in vivo, where the three Fgfrs differentially control the normal generation of OLPs from the embryonic ventral forebrain.


Subject(s)
Embryonic Stem Cells/physiology , Neurogenesis/genetics , Oligodendroglia/physiology , Prosencephalon/embryology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 2/physiology , Signal Transduction/genetics , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblast Growth Factors/physiology , Mice , Mice, Knockout , Mice, Transgenic , Oligodendroglia/cytology , Prosencephalon/cytology , Prosencephalon/physiology , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptor, Fibroblast Growth Factor, Type 2/genetics , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/physiology
18.
Cancer Res ; 70(20): 8211-21, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20924106

ABSTRACT

Vascular endothelial growth factor and its receptors, including Flt-1 and Flk-1, are involved in angiogenesis under physiologic and pathologic conditions. Recently, Flt-1-expressing cells were reported to contribute to the intracranial growth of glioma cells. However, the role of Flt-1 signaling in solid tumor growth in s.c. tissue has not been elucidated. To investigate how Flt-1 signaling is involved in the proliferation of solid tumors, we implanted tumor cells into wild-type (Wt) and Flt-1 tyrosine kinase (TK)-deficient (Flt-1 TK(-/-)) mice. Growth of HSML and B16 but not Lewis lung carcinoma cell in s.c. tissue was significantly decreased in Flt-1 TK(-/-) mice. Angiogenesis in HSML and B16 tumors was remarkably reduced in Flt-1 TK(-/-) mice. Moreover, the infiltration of macrophage lineage cells into HSML and B16 tumors was clearly suppressed in Flt-1 TK(-/-) mice. Pericyte marker(+) cells were also reduced in Flt-1 TK(-/-) mice. However, in the border area of tumor, angiogenesis and the infiltration of macrophage lineage cell were basically similar between Wt and Flt-1 TK(-/-) mice. In bone marrow (BM) transplantation experiments, tumor angiogenesis, infiltration of macrophage lineage cells, and tumor growth were significantly suppressed in Wt/Flt-1 TK(-/-) mice implanted with Flt-1 TK(-/-) BM cells compared with those implanted with Wt BM cells. We conclude that Flt-1 signaling is involved in the function of BM-derived cell, such as the migration of macrophages into cancerous tissues, and significantly contributes to angiogenesis and tumor progression.


Subject(s)
Bone Marrow Cells/physiology , Macrophages/physiology , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor Receptor-1/physiology , Alternative Splicing , Animals , Bone Marrow Cells/pathology , Carcinoma, Lewis Lung/pathology , Cell Division , Cell Line, Tumor , DNA Primers , Female , Fibroblast Growth Factor 2/genetics , Lung Neoplasms/pathology , Macrophages/pathology , Mice , Mice, Knockout , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Uterine Neoplasms/pathology , Vascular Endothelial Growth Factor A/genetics
19.
J Neurosci ; 30(19): 6595-606, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20463222

ABSTRACT

Identification of intracellular signaling pathways necessary for appropriate axon guidance is challenging because many CNS populations used to study these events contain multiple cell types. Here, we resolve this issue by using mouse embryonic stem (ES) cells that were directed to differentiate into a population of motoneurons that exclusively innervate epaxial muscles [medial median motor column (MMCm) motoneurons]. These ES cell-derived MMCm motoneurons, like their endogenous counterparts, express fibroblast growth factor receptor 1 (FGFR1) and selectively extend axons toward the epaxial trophin FGF8. Unlike wild-type MMCm motoneurons, FGFR1(-/-) MMCm motoneurons show guidance defects when transplanted into the neural tube of chick embryos. Furthermore, activation of FGFR1 selectively signals through mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) for appropriate guidance in vitro, whereas overexpression of constitutively active MAPK/ERK in transplanted, or endogenous chick, MMCm cells causes guidance defects in vivo. These results indicate that MAPK/ERK activation downstream of FGFR1 is necessary for MMCm motor axon guidance and that ES cell-derived neurons provide an important tool for dissecting intracellular pathways required for axon guidance.


Subject(s)
Axons/physiology , Cell Movement/physiology , MAP Kinase Signaling System , Motor Neurons/physiology , Posture/physiology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Cells, Cultured , Chick Embryo , Embryonic Stem Cells , Extracellular Signal-Regulated MAP Kinases/metabolism , Extremities/growth & development , Extremities/innervation , Extremities/physiology , Fibroblast Growth Factor 8/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Neural Tube/growth & development , Neural Tube/physiology , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Stem Cell Transplantation
20.
J Neurosci ; 29(46): 14571-80, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19923290

ABSTRACT

During corticogenesis, the balance between the self-renewal of radial glial stem cells and the production of their descendent progenitor cells is essential in generating the correct size and cell composition of the neocortex. How the stem-to-progenitor cell transition is regulated is poorly understood. FGFs are commonly implicated in promoting proliferation of neural precursor cells, but it is unclear how they exert their effects on stem cells, progenitor cells, or both in vivo. Here, three FGF receptor genes are simultaneously deleted during cortical neurogenesis. In these mutants, radial glia are depleted due to an increased transition from an uncommitted state to a more differentiated one, initially causing an increase in progenitors, but ultimately resulting in a smaller cortex. The proliferation rate of progenitors themselves, however, is unchanged. These results indicate that FGFs normally repress the radial glia to progenitor cell transition during corticogenesis.


Subject(s)
Cerebral Cortex/physiology , Neurogenesis , Neuroglia/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 2/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Signal Transduction , Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation , Cell Survival/genetics , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Female , Mice , Mice, Knockout , Mice, Transgenic , Neurogenesis/genetics , Neuroglia/cytology , Pregnancy , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 3/deficiency , Receptor, Fibroblast Growth Factor, Type 3/genetics , Signal Transduction/genetics , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...