Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502405

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of the central nervous system (CNS). MS commonly affects the cerebellum causing acute and chronic symptoms. Cerebellar signs significantly contribute to clinical disability, and symptoms such as tremor, ataxia, and dysarthria are difficult to treat. Fibroblast growth factors (FGFs) and their receptors (FGFRs) are involved in demyelinating pathologies such as MS. In autopsy tissue from patients with MS, increased expression of FGF1, FGF2, FGF9, and FGFR1 was found in lesion areas. Recent research using mouse models has focused on regions such as the spinal cord, and data on the expression of FGF/FGFR in the cerebellum are not available. In recent EAE studies, we detected that oligodendrocyte-specific deletion of FGFRs results in a milder disease course, less cellular infiltrates, and reduced neurodegeneration in the spinal cord. The objective of this study was to characterize the role of FGFR1 in oligodendrocytes in the cerebellum. Conditional deletion of FGFR1 in oligodendrocytes (Fgfr1ind-/-) was achieved by tamoxifen application, EAE was induced using the MOG35-55 peptide. The cerebellum was analyzed by histology, immunohistochemistry, and western blot. At day 62 p.i., Fgfr1ind-/- mice showed less myelin and axonal degeneration compared to FGFR1-competent mice. Infiltration of CD3(+) T cells, Mac3(+) cells, B220(+) B cells and IgG(+) plasma cells in cerebellar white matter lesions (WML) was less in Fgfr1ind-/-mice. There were no effects on the number of OPC or mature oligodendrocytes in white matter lesion (WML). Expression of FGF2 and FGF9 associated with less myelin and axonal degeneration, and of the pro-inflammatory cytokines IL-1ß, IL-6, and CD200 was downregulated in Fgfr1ind-/- mice. The FGF/FGFR signaling protein pAkt, BDNF, and TrkB were increased in Fgfr1ind-/- mice. These data suggest that cell-specific deletion of FGFR1 in oligodendrocytes has anti-inflammatory and neuroprotective effects in the cerebellum in the EAE disease model of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Oligodendroglia/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Cerebellum/metabolism , Cerebellum/pathology , Demyelinating Diseases/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Fibroblast Growth Factors/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/metabolism , Multiple Sclerosis/physiopathology , Myelin Sheath/metabolism , Myelin-Oligodendrocyte Glycoprotein/pharmacology , Oligodendroglia/pathology , Phosphorylation , Receptor, Fibroblast Growth Factor, Type 1/physiology , Signal Transduction/drug effects , Spinal Cord/metabolism , White Matter/metabolism
2.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946884

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive breast tumor subtype characterized by poor clinical outcome. In recent years, numerous advancements have been made to better understand the biological landscape of TNBC, though appropriate targets still remain to be determined. In the present study, we have determined that the expression levels of FGF2 and S100A4 are higher in TNBC with respect to non-TNBC patients when analyzing "The Invasive Breast Cancer Cohort of The Cancer Genome Atlas" (TCGA) dataset. In addition, we have found that the gene expression of FGF2 is positively correlated with S100A4 in TNBC samples. Performing quantitative PCR, Western blot, CRISPR/Cas9 genome editing, promoter studies, immunofluorescence analysis, subcellular fractionation studies, and ChIP assays, we have also demonstrated that FGF2 induces in TNBC cells the upregulation and secretion of S100A4 via FGFR1, along with the ERK1/2-AKT-c-Rel transduction signaling. Using conditioned medium from TNBC cells stimulated with FGF2, we have also ascertained that the paracrine activation of the S100A4/RAGE pathway triggers angiogenic effects in vascular endothelial cells (HUVECs) and promotes the migration of cancer-associated fibroblasts (CAFs). Collectively, our data provide novel insights into the action of the FGF2/FGFR1 axis through S100A4 toward stimulatory effects elicited in TNBC cells.


Subject(s)
Fibroblast Growth Factor 2/physiology , Neoplasm Proteins/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , S100 Calcium-Binding Protein A4/physiology , Signal Transduction/physiology , Triple Negative Breast Neoplasms/physiopathology , Antigens, Neoplasm/physiology , Cell Movement/drug effects , Culture Media, Conditioned/pharmacology , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic/physiology , Human Umbilical Vein Endothelial Cells , Humans , Mitogen-Activated Protein Kinases/physiology , Neovascularization, Pathologic/physiopathology , Paracrine Communication , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-rel/physiology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/blood supply , Tumor Cells, Cultured
3.
Clin Cancer Res ; 27(15): 4379-4396, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34011560

ABSTRACT

PURPOSE: FGFR1 overexpression has been associated with endocrine resistance in ER+ breast cancer. We found FGFR1 localized in the nucleus of breast cancer cells in primary tumors resistant to estrogen suppression. We investigated a role of nuclear FGFR1 on gene transcription and antiestrogen resistance. EXPERIMENTAL DESIGN: Tumors from patients treated with letrozole were subjected to Ki67 and FGFR1 IHC. MCF7 cells were transduced with FGFR1(SP-)(NLS) to promote nuclear FGFR1 overexpression. FGFR1 genomic activity in ER+/FGFR1-amplified breast cancer cells ± FOXA1 siRNA or ± the FGFR tyrosine kinase inhibitor (TKI) erdafitinib was examined by chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-seq). The nuclear and chromatin-bound FGFR1 interactome was investigated by mass spectrometry (MS). RESULTS: High nuclear FGFR1 expression in ER+ primary tumors positively correlated with post-letrozole Ki67 values. Nuclear FGFR1 overexpression influenced gene transcription and promoted resistance to estrogen suppression and to fulvestrant in vivo. A gene expression signature induced by nuclear FGFR1 correlated with shorter survival in the METABRIC cohort of patients treated with antiestrogens. ChIP-Seq revealed FGFR1 occupancy at transcription start sites, overlapping with active transcription histone marks. MS analysis of the nuclear FGFR1 interactome identified phosphorylated RNA-Polymerase II and FOXA1, with FOXA1 RNAi impairing FGFR1 recruitment to chromatin. Treatment with erdafitinib did not impair nuclear FGFR1 translocation and genomic activity. CONCLUSIONS: These data suggest nuclear FGFR1 contributes to endocrine resistance by modulating gene transcription in ER+ breast cancer. Nuclear FGFR1 activity was unaffected by FGFR TKIs, thus supporting the development of treatment strategies to inhibit nuclear FGFR1 in ER+/FGFR1 overexpressing breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Estrogen Receptor Modulators/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/physiology , Transcription, Genetic/physiology , Breast Neoplasms/chemistry , Cell Nucleus , Female , Humans , Receptors, Estrogen/analysis , Tumor Cells, Cultured
4.
Transl Res ; 235: 129-143, 2021 09.
Article in English | MEDLINE | ID: mdl-33894400

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is a genetic disease characterized by vascular dysplasia. Mutations of the endoglin (ENG) gene that encodes a co-receptor of the transforming growth factor ß1 signaling pathway cause type I HHT. ENG is primarily expressed in endothelial cells (ECs), but its interaction with other key angiogenic pathways to control angiogenesis has not been well addressed. The aim of this study is to investigate ENG interplay with VEGFR2, FGFR1 and TIE2 in primary human ECs. ENG was knocked-down with siRNA in human umbilical vein ECs (HUVECs) and human lung microvascular ECs (HMVEC-L). Gene expression was measured by RT-qPCR and Western blotting. Cell signaling pathway activation was analyzed by detecting phosphor-ERK and phosphor-AKT levels. Cell migration and apoptosis were assessed using the Boyden chamber assay and the CCK-8 Kit, respectively. Loss of ENG in HUVECs led to significantly reduced expression of VEGFR2 but not TIE2 or FGFR1, which was also confirmed in HMVEC-L. HUVECs lacking ENG had significantly lower levels of active Rac1 and a substantial reduction of the transcription factor Sp1, an activator of VEGFR2 transcription, in nuclei. Furthermore, VEGF- but not bFGF- or angiopoietin-1-induced phosphor-ERK and phosphor-AKT were suppressed in ENG deficient HUVECs. Functional analysis revealed that ENG knockdown inhibited cell migratory but enhanced anti-apoptotic activity induced by VEGF. In contrast, bFGF, angiopoietin-1 and -2 induced HUVEC migration and anti-apoptotic activities were not affected by ENG knockdown. In conclusion, ENG deficiency alters the VEGF/VEGFR2 pathway, which may play a role in HHT pathogenesis.


Subject(s)
Endoglin/physiology , Endothelial Cells/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, TIE-2/physiology , Telangiectasia, Hereditary Hemorrhagic/etiology , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/physiology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Proto-Oncogene Proteins c-akt/physiology
5.
Exp Cell Res ; 400(2): 112490, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33484747

ABSTRACT

Tumor neovascularization may occur via both angiogenic and vasculogenic events. In order to investigate the vessel formation during tumor growth, we developed a novel experimental model that takes into account the differentiative and tumorigenic properties of Embryonic Stem cells (ESCs). Leukemia Inhibitory Factor-deprived murine ESCs were grafted on the top of the chick embryo chorionallantoic membrane (CAM) in ovo. Cell grafts progressively grew, forming a vascularized mass within 10 days. At this stage, the grafts are formed by cells with differentiative features representative of all three germ layers, thus originating teratomas, a germinal cell tumor. In addition, ESC supports neovascular events by recruiting host capillaries from surrounding tissue that infiltrates the tumor mass. Moreover, immunofluorescence studies demonstrate that perfused active blood vessels within the tumor are of both avian and murine origin because of the simultaneous occurrence of angiogenic and vasculogenic events. In conclusion, the chick embryo ESC/CAM-derived teratoma model may represent a useful approach to investigate both vasculogenic and angiogenic events during tumor growth and for the study of natural and synthetic modulators of the two processes.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/pathology , Neovascularization, Pathologic , Receptor, Fibroblast Growth Factor, Type 1/physiology , Teratoma/blood supply , Teratoma/pathology , Animals , Chick Embryo , Chorioallantoic Membrane , Embryonic Stem Cells/metabolism , Mice , Mice, Knockout , Teratoma/metabolism
6.
Neuropharmacology ; 170: 108070, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32229140

ABSTRACT

The majority of the fibroblast growth factor receptor 1-serotonin 1 A receptor (FGFR1-5-HT1AR) heterocomplexes in the hippocampus appeared to be located mainly in the neuronal networks and a relevant target for antidepressant drugs. Through a neurochemical and electrophysiological analysis it was therefore tested in the current study if astrocytic FGFR1-5-HT1AR heterocomplexes also exist in hippocampus. They may modulate the structure and function of astroglia in the hippocampus leading to possible changes in the gamma oscillations. Localization of hippocampal FGFR1-5-HT1AR heterocomplexes in astrocytes was found using in situ proximity ligation assay combined with immunohistochemistry using glial fibrillary acidic protein (GFAP) immunoreactivity as a marker for astroglia. Acute i.c.v. treatment with 8-OH-DPAT alone or together with basic fibroblast growth factor (FGF2) significantly increased FGFR1-5-HT1AR heterocomplexes in the GFAP positive cells, especially in the polymorphic layer of the dentate gyrus (PoDG) but also in the CA3 area upon combined treatment. No other hippocampal regions were studied. Also, structural plasticity changes were observed in the astrocytes, especially in the PoDG region, upon these pharmacological treatments. They may also be of relevance for enhancing the astroglial volume transmission with increased modulation of the neuronal networks in the regions studied. The effects of combined FGF2 and 5-HT agonist treatments on gamma oscillations point to a significant antagonistic interaction in astroglial FGFR1-5-HT1AR heterocomplexes that may contribute to counteraction of the 5-HT1AR-mediated decrease of gamma oscillations. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.


Subject(s)
Astrocytes/physiology , Fibroblast Growth Factor 2/physiology , Gamma Rhythm/physiology , Hippocampus/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Serotonin, 5-HT1A/physiology , Serotonin/physiology , Animals , Astrocytes/drug effects , Gamma Rhythm/drug effects , Hippocampus/drug effects , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/agonists , Serotonin 5-HT1 Receptor Agonists/pharmacology
7.
Diabetes ; 69(7): 1368-1381, 2020 07.
Article in English | MEDLINE | ID: mdl-32332158

ABSTRACT

α-Klotho is a circulating factor with well-documented antiaging properties. However, the central role of α-klotho in metabolism remains largely unexplored. The current study investigated the potential role of central α-klotho to modulate neuropeptide Y/agouti-related peptide (NPY/AgRP)-expressing neurons, energy balance, and glucose homeostasis. Intracerebroventricular administration of α-klotho suppressed food intake, improved glucose profiles, and reduced body weight in mouse models of type 1 and 2 diabetes. Furthermore, central α-klotho inhibition via an anti-α-klotho antibody impaired glucose tolerance. Ex vivo patch clamp electrophysiology and immunohistochemical analysis revealed that α-klotho suppresses NPY/AgRP neuron activity, at least in part, by enhancing miniature inhibitory postsynaptic currents. Experiments in hypothalamic GT1-7 cells observed that α-klotho induces phosphorylation of AKTser473, ERKthr202/tyr204, and FOXO1ser256 as well as blunts AgRP gene transcription. Mechanistically, fibroblast growth factor receptor 1 (FGFR1) inhibition abolished the downstream signaling of α-klotho, negated its ability to modulate NPY/AgRP neurons, and blunted its therapeutic effects. Phosphatidylinositol 3 kinase (PI3K) inhibition also abolished α-klotho's ability to suppress food intake and improve glucose clearance. These results indicate a prominent role of hypothalamic α-klotho/FGFR1/PI3K signaling in the modulation of NPY/AgRP neuron activity and maintenance of energy homeostasis, thus providing new insight into the pathophysiology of metabolic disease.


Subject(s)
Agouti-Related Protein/physiology , Glucuronidase/physiology , Neurons/physiology , Neuropeptide Y/physiology , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/drug therapy , Energy Metabolism , Glucose/metabolism , Glucuronidase/administration & dosage , Infusions, Intraventricular , Klotho Proteins , Male , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Signal Transduction/physiology
8.
Front Endocrinol (Lausanne) ; 11: 588459, 2020.
Article in English | MEDLINE | ID: mdl-33679600

ABSTRACT

In humans and mice, inactivating mutations in fibroblast growth factor receptor 1 (Fgfr1) lead to gonadotropin-releasing hormone (GnRH) deficiency and a host of downstream reproductive disorders. It was unclear if Fgfr1 signaling directly upon GnRH neurons critically drove the establishment of a functional GnRH system. To answer this question, we generated a mouse model with a conditional deletion of Fgfr1 in GnRH neurons using the Cre/loxP approach. These mice, called Fgfr1cKO mice, were examined along with control mice for their pubertal onset and a host of reproductive axis functions. Our results showed that Fgfr1cKO mice harbored no detectable defects in the GnRH system and pubertal onset, suffered only subtle changes in the pituitary function, but exhibited significantly disrupted testicular and ovarian morphology at 25 days of age, indicating impaired gametogenesis at a young age. However, these disruptions were transient and became undetectable in older mice. Our results suggest that Fgfr1 signaling directly on GnRH neurons supports, to some extent, the reproductive axis function in the period leading to the early phase of puberty, but is not critically required for pubertal onset or reproductive maintenance in sexually mature animals.


Subject(s)
Gametogenesis , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Receptor, Fibroblast Growth Factor, Type 1/physiology , Reproduction , Sexual Maturation , Animals , Female , Integrases , Male , Mice , Mice, Knockout , Mutation , Signal Transduction
9.
Cells ; 8(7)2019 07 13.
Article in English | MEDLINE | ID: mdl-31337028

ABSTRACT

Glioblastoma is the most lethal brain cancer in adults, with no known cure. This cancer is characterized by a pronounced genetic heterogeneity, but aberrant activation of receptor tyrosine kinase signaling is among the most frequent molecular alterations in glioblastoma. Somatic mutations of fibroblast growth factor receptors (FGFRs) are rare in these cancers, but many studies have documented that signaling through FGFRs impacts glioblastoma progression and patient survival. Small-molecule inhibitors of FGFR tyrosine kinases are currently being trialed, underlining the therapeutic potential of blocking this signaling pathway. Nevertheless, a comprehensive overview of the state of the art of the literature on FGFRs in glioblastoma is lacking. Here, we review the evidence for the biological functions of FGFRs in glioblastoma, as well as pharmacological approaches to targeting these receptors.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 2/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Receptor, Fibroblast Growth Factor, Type 4/physiology , Disease Progression , Humans , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 2/chemistry , Receptor, Fibroblast Growth Factor, Type 3/chemistry , Receptor, Fibroblast Growth Factor, Type 4/chemistry
10.
Curr Pharm Des ; 25(23): 2555-2568, 2019.
Article in English | MEDLINE | ID: mdl-31333086

ABSTRACT

Neurodegenerative disorders are the most devastating disorder of the nervous system. The pathological basis of neurodegeneration is linked with dysfunctional protein trafficking, mitochondrial stress, environmental factors and aging. With the identification of insulin and insulin receptors in some parts of the brain, it has become evident that certain metabolic conditions associated with insulin dysfunction like Type 2 diabetes mellitus (T2DM), dyslipidemia, obesity etc., are also known to contribute to neurodegeneration mainly Alzheimer's Disease (AD). Recently, a member of the fibroblast growth factor (FGF) superfamily, FGF21 has proved tremendous efficacy in diseases like diabetes mellitus, obesity and insulin resistance (IR). Increased levels of FGF21 have been reported to exert multiple beneficial effects in metabolic syndrome. FGF21 receptors are present in certain areas of the brain involved in learning and memory. However, despite extensive research, its function as a neuroprotectant in AD remains elusive. FGF21 is a circulating endocrine hormone which is mainly secreted by the liver primarily in fasting conditions. FGF21 exerts its effects after binding to FGFR1 and co-receptor, ß-klotho (KLB). It is involved in regulating energy via glucose and lipid metabolism. It is believed that aberrant FGF21 signalling might account for various anomalies like neurodegeneration, cancer, metabolic dysfunction etc. Hence, this review will majorly focus on FGF21 role as a neuroprotectant and potential metabolic regulator. Moreover, we will also review its potential as an emerging candidate for combating metabolic stress induced neurodegenerative abnormalities.


Subject(s)
Alzheimer Disease/therapy , Fibroblast Growth Factors/physiology , Neuroprotection , Humans , Klotho Proteins , Membrane Proteins/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology
11.
Neuropharmacology ; 152: 67-77, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30445101

ABSTRACT

Neuronal events are regulated by the integration of several complex signaling networks in which G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs) are considered key players of an intense bidirectional cross-communication in the cell, generating signaling mechanisms that, at the same time, connect and diversify the traditional signal transduction pathways activated by the single receptor. For this receptor-receptor crosstalk, the two classes of receptors form heteroreceptor complexes resulting in RTKs transactivation and in growth-promoting signals. In this review, we describe heteroreceptor complexes between GPCR and RTKs in the central nervous system (CNS) and their functional effects in controlling a variety of neuronal effects, ranging from development, proliferation, differentiation and migration, to survival, repair, synaptic transmission and plasticity. In this interaction, RTKs can also recruit components of the G protein signaling cascade, creating a bidirectional intricate interplay that provides complex control over multiple cellular events. These heteroreceptor complexes, by the integration of different signals, have recently attracted a growing interest as novel molecular target for depressive disorders. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.


Subject(s)
Receptor Cross-Talk/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Brain/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/physiology , Signal Transduction/physiology
12.
Proc Natl Acad Sci U S A ; 115(33): 8388-8393, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061390

ABSTRACT

The mechanosensory hair cells of the inner ear are required for hearing and balance and have a distinctive apical structure, the hair bundle, that converts mechanical stimuli into electrical signals. This structure comprises a single cilium, the kinocilium, lying adjacent to an ensemble of actin-based projections known as stereocilia. Hair bundle polarity depends on kinociliary protocadherin-15 (Pcdh15) localization. Protocadherin-15 is found only in hair-cell kinocilia, and is not localized to the primary cilia of adjacent supporting cells. Thus, Pcdh15 must be specifically targeted and trafficked into the hair-cell kinocilium. Here we show that kinocilial Pcdh15 trafficking relies on cell type-specific coupling to the generic intraflagellar transport (IFT) transport mechanism. We uncover a role for fibroblast growth factor receptor 1 (FGFR1) in loading Pcdh15 onto kinociliary transport particles in hair cells. We find that on activation, FGFR1 binds and phosphorylates Pcdh15. Moreover, we find a previously uncharacterized role for clathrin in coupling this kinocilia-specific cargo with the anterograde IFT-B complex through the adaptor, DAB2. Our results identify a modified ciliary transport pathway used for Pcdh15 transport into the cilium of the inner ear hair cell and coordinated by FGFR1 activity.


Subject(s)
Cadherins/physiology , Flagella/metabolism , Hair Cells, Auditory, Inner/metabolism , Protein Precursors/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/physiology , Animals , Apoptosis Regulatory Proteins , Cadherin Related Proteins , Chick Embryo , Clathrin/physiology , Mice , Phosphorylation , Protein Transport , Receptor, Fibroblast Growth Factor, Type 1/analysis
13.
Cell Mol Life Sci ; 75(19): 3649-3661, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29705951

ABSTRACT

Axons sense molecular cues in their environment to arrive at their post-synaptic targets. While many of the molecular cues have been identified, the mechanisms that regulate their spatiotemporal expression remain elusive. We examined here the transcriptional regulation of the guidance gene slit1 both in vitro and in vivo by specific fibroblast growth factor receptors (Fgfrs). We identified an Fgf-responsive 2.3 kb slit1 promoter sequence that recapitulates spatiotemporal endogenous expression in the neural tube and eye of Xenopus embryos. We found that signaling through Fgfr1 is the main regulator of slit1 expression both in vitro in A6 kidney epithelial cells, and in the Xenopus forebrain, even when other Fgfr subtypes are present in cells. These data argue that a specific signaling pathway downstream of Fgfr1 controls in a cell-autonomous manner slit1 forebrain expression and are novel in identifying a specific growth factor receptor for in vivo control of the expression of a key embryonic axon guidance cue.


Subject(s)
Axon Guidance/genetics , Nerve Tissue Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/physiology , Xenopus Proteins/genetics , Animals , Cells, Cultured , Embryo, Nonmammalian , Female , Gene Expression Regulation, Developmental , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/physiology , Transcriptional Activation/physiology , Xenopus laevis
14.
Toxicol Pathol ; 45(7): 904-910, 2017 10.
Article in English | MEDLINE | ID: mdl-29096595

ABSTRACT

Fibroblast growth factor-23 (FGF23) is a bone-derived hormone, mainly produced by osteoblasts and osteocytes in response to increased extracellular phosphate and circulating vitamin D hormone. Endocrine FGF23 signaling requires co-expression of the ubiquitously expressed FGF receptor 1 (FGFR1) and the co-receptor α-Klotho (Klotho). In proximal renal tubules, FGF23 suppresses the membrane expression of the sodium-phosphate cotransporters Npt2a and Npt2c which mediate urinary reabsorption of filtered phosphate. In addition, FGF23 suppresses proximal tubular expression of 1α-hydroxylase, the key enzyme responsible for vitamin D hormone production. In distal renal tubules, FGF23 signaling activates with-no-lysine kinase 4, leading to increased renal tubular reabsorption of calcium and sodium. Therefore, FGF23 is not only a phosphaturic but also a calcium- and sodium-conserving hormone, a finding that may have important implications for the pathophysiology of chronic kidney disease. Besides these endocrine, Klotho-dependent functions of FGF23, FGF23 is also an auto-/paracrine suppressor of tissue-nonspecific alkaline phosphatase transcription via Klotho-independent FGFR3 signaling, leading to local inhibition of mineralization through accumulation of pyrophosphate. In addition, FGF23 may target the heart via an FGFR4-mediated Klotho-independent signaling cascade. Taken together, there is emerging evidence that FGF23 is a pleiotropic hormone, linking bone with several other organ systems.


Subject(s)
Bone and Bones/physiology , Fibroblast Growth Factors/physiology , Glucuronidase/physiology , Autocrine Communication , Calcification, Physiologic , Cardiovascular System , Fibroblast Growth Factor-23 , Humans , Immunomodulation , Kidney Tubules, Proximal/physiology , Klotho Proteins , Paracrine Communication , Phosphates/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Receptor, Fibroblast Growth Factor, Type 4/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIa/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIc/physiology
15.
Endocrinology ; 158(10): 3081-3082, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28977611

ABSTRACT

Currently there are few, if any, approved and effective medicines for the attenuation of obesity, diabetes, and insulin resistance. This commentary addresses a communication describing the effect on glucose dynamics and obesity of a peptide monoclonal antibody for fibroblast growth factor (FGF) receptor 1c isoform (FGFR1c). The general lack of suitable, effective drugs is discussed, as is the treatment potential of FGF family receptors. The FGFR1c monoclonal antibody developed by the authors lowers body weight gain, blood glucose, and adipose tissue weight. It also enhances glucose uptake by fat cells [white adipose tissue (WAT) and 3T3-L1]. The robust weight loss, fat loss, and lower blood glucose were attributed to an observed potential futile cycle of continuous lipogenesis and lipolysis and adenosine triphosphate use in WAT.


Subject(s)
Antibodies, Monoclonal/pharmacology , Obesity/metabolism , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , 3T3-L1 Cells , Adenosine Triphosphate/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Antibodies, Monoclonal/therapeutic use , Blood Glucose/analysis , Blood Glucose/drug effects , Glucose/metabolism , Humans , Lipogenesis/drug effects , Lipolysis/drug effects , Mice , Obesity/drug therapy , Obesity/pathology , Organ Size/drug effects , Receptor, Fibroblast Growth Factor, Type 1/immunology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Weight Gain/drug effects
16.
Oncogene ; 36(48): 6668-6679, 2017 11 30.
Article in English | MEDLINE | ID: mdl-28783178

ABSTRACT

Epidemiological evidence implicates excess adipose tissue in increasing cancer risk. Despite a steeply rising global prevalence of obesity, how adiposity contributes to transformation (stage a non-tumorigenic cell undergoes to become malignant) is unknown. To determine the factors in adipose tissue that stimulate transformation, we used a novel ex vivo system of visceral adipose tissue (VAT)-condition medium-stimulated epithelial cell growth in soft agar. To extend this system in vivo, we used a murine lipectomy model of ultraviolet light B-induced, VAT-promoted skin tumor formation. We found that VAT from mice and obese human donors stimulated growth in soft agar of non-tumorigenic epithelial cells. The difference in VAT activity was associated with fibroblast growth factor-2 (FGF2) levels. Moreover, human and mouse VAT failed to stimulate growth in soft of agar in cells deficient in FGFR-1 (FGF2 receptor). We also demonstrated that circulating levels of FGF2 were associated with non-melanoma tumor formation in vivo. These data implicate FGF2 as a major factor VAT releases to transform epithelial cells-a novel, potential pathway of VAT-enhanced tumorigenesis. Strategies designed to deplete VAT stores of FGF2 or inhibit FGFR-1 in abdominally obese individuals may be important cancer prevention strategies as well as adjuvant therapies for improving outcomes.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Receptor, Fibroblast Growth Factor, Type 1/physiology , Animals , Cell Line , Diet, High-Fat/adverse effects , Female , Fibroblast Growth Factor 2/physiology , Humans , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Mice, Inbred Strains , Signal Transduction
17.
Dev Cell ; 41(5): 496-510.e5, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28552559

ABSTRACT

Fibroblast growth factor 4 (FGF4) is the key signal driving specification of primitive endoderm (PrE) versus pluripotent epiblast (EPI) within the inner cell mass (ICM) of the mouse blastocyst. To gain insight into the receptor(s) responding to FGF4 within ICM cells, we combined single-cell-resolution quantitative imaging with single-cell transcriptomics of wild-type and Fgf receptor (Fgfr) mutant embryos. Despite the PrE-specific expression of FGFR2, it is FGFR1, expressed by all ICM cells, that is critical for establishment of a PrE identity. Signaling through FGFR1 is also required to constrain levels of the pluripotency-associated factor NANOG in EPI cells. However, the activity of both receptors is required for lineage establishment within the ICM. Gene expression profiling of 534 single ICM cells identified distinct downstream targets associated with each receptor. These data lead us to propose a model whereby unique and additive activities of FGFR1 and FGFR2 within the ICM coordinate establishment of two distinct lineages.


Subject(s)
Blastocyst Inner Cell Mass/cytology , Cell Lineage/genetics , Embryo, Mammalian/cytology , Endoderm/cytology , Fibroblast Growth Factor 4/metabolism , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 2/physiology , Animals , Blastocyst Inner Cell Mass/metabolism , Cells, Cultured , Embryo, Mammalian/metabolism , Endoderm/metabolism , Female , Humans , Mice , Mice, Knockout
18.
Dev Cell ; 41(5): 511-526.e4, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28552557

ABSTRACT

Activation of the FGF signaling pathway during preimplantation development of the mouse embryo is known to be essential for differentiation of the inner cell mass and the formation of the primitive endoderm (PrE). We now show using fluorescent reporter knockin lines that Fgfr1 is expressed in all cell populations of the blastocyst, while Fgfr2 expression becomes restricted to extraembryonic lineages, including the PrE. We further show that loss of both receptors prevents the development of the PrE and demonstrate that FGFR1 plays a more prominent role in this process than FGFR2. Finally, we document an essential role for FGFRs in embryonic stem cell (ESC) differentiation, with FGFR1 again having a greater influence than FGFR2 in ESC exit from the pluripotent state. Collectively, these results identify mechanisms through which FGF signaling regulates inner cell mass lineage restriction and cell commitment during preimplantation development.


Subject(s)
Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Endoderm/cytology , Fibroblast Growth Factor 4/metabolism , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 2/physiology , Animals , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Endoderm/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
19.
Oncotarget ; 7(52): 87124-87135, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27893433

ABSTRACT

Recent evidence suggests that T-box transcription factor brachyury plays an important role in lung cancer development and progression. However, the mechanisms underlying brachyury-driven cellular processes remain unclear. Here we found that fibroblast growth factor receptor 1/mitogen-activated protein kinase (FGFR1/MAPK) signaling regulated brachyury in lung cancer. Analysis of FGFR1-4 and brachyury expression in human lung tumor tissue and cell lines found that only expression of FGFR1 was positively correlated with brachyury expression. Specific knockdown of FGFR1 by siRNA suppressed brachyury expression and epithelial-mesenchymal transition (EMT) (upregulation of E-cadherin and ß-catenin and downregulation of Snail and fibronectin), whereas forced overexpression of FGFR1 induced brachyury expression and promoted EMT in lung cancer cells. Activation of fibroblast growth factor (FGF)/FGFR1 signaling promoted phosphorylated MAPK extracellular signal-regulated kinase (ERK) 1/2 translocation from cytoplasm to nucleus, upregulated brachyury expression, and increased cell growth and invasion. In addition, human lung cancer cells with higher brachyury expression were more sensitive to inhibitors targeting FGFR1/MAPK pathway. These findings suggest that FGFR1/MAPK may be important for brachyury activation in lung cancer, and this pathway may be an appealing therapeutic target for a subset of brachyury-driven lung cancer.


Subject(s)
Fetal Proteins/physiology , Lung Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 1/physiology , T-Box Domain Proteins/physiology , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Humans , MAP Kinase Signaling System/physiology , Neoplasm Invasiveness , Receptor, Fibroblast Growth Factor, Type 1/analysis , Signal Transduction/physiology
20.
Mol Cell Biochem ; 411(1-2): 289-98, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26476925

ABSTRACT

The migration and proliferation of EPCs are crucial for re-endothelialization in vascular repair and development. Id1 has a regulatory role in the regulation of EPCs migration and proliferation. Based on these findings, we hypothesized that Id1 plays a regulatory role in modulating the migration and proliferation of EPCs by interaction with other factors. Herein, we report that the Id1 protein and E-box protein E2-2 regulate EPCs function with completely opposite effects. Id1 plays a positive role in the regulation of EPC proliferation and migration, while endogenous E2-2 appears to be a negative regulator. Immunoprecipitation and immunofluorescence assay revealed that the Id1 protein interacts and co-localizes with the E2-2 protein in EPCs. Further, endogenous E2-2 protein was found to block EPCs function via the inhibition of FGFR1 and VEGFR2 expression. The overexpression and silencing of Id1 have no direct regulatory role on VEGFR2 and FGFR1 expression. On the other hand, Id1 relieves the E2-2-mediated repression of FGFR1 and VEGFR2 expression to modulate EPCs proliferation, migration, and tube formation in vitro. In summary, we demonstrated that Id1 and E2-2 are critical regulators of EPCs function in vitro. Id1 interacts with E2-2 and relieves the E2-2-mediated repression of FGFR1 and VEGFR2 expression to modulate EPCs functions. Id1 and E2-2 may represent novel therapeutic targets for re-endothelialization in vascular damage and repair.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Endothelial Progenitor Cells/cytology , Inhibitor of Differentiation Protein 1/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology , Animals , Cell Movement/physiology , Cell Proliferation/physiology , Male , Mice , Transcription Factor 4
SELECTION OF CITATIONS
SEARCH DETAIL