Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Nat Neurosci ; 23(10): 1297-1306, 2020 10.
Article in English | MEDLINE | ID: mdl-32895565

ABSTRACT

How astrocytes grow and integrate into neural circuits remains poorly defined. Zebrafish are well suited for such investigations, but bona fide astrocytes have not been described in this system. Here we characterize a zebrafish cell type that is remarkably similar to mammalian astrocytes that derive from radial glial cells and elaborate processes to establish their territories at early larval stages. Zebrafish astrocytes associate closely with synapses, tile with one another and express markers, including Glast and glutamine synthetase. Once integrated into circuits, they exhibit whole-cell and microdomain Ca2+ transients, which are sensitive to norepinephrine. Finally, using a cell-specific CRISPR-Cas9 approach, we demonstrate that fgfr3 and fgfr4 are required for vertebrate astrocyte morphogenesis. This work provides the first visualization of astrocyte morphogenesis from stem cell to post-mitotic astrocyte in vivo, identifies a role for Fgf receptors in vertebrate astrocytes and establishes zebrafish as a valuable new model system to study astrocyte biology in vivo.


Subject(s)
Astrocytes/physiology , Brain/growth & development , Ependymoglial Cells/physiology , Morphogenesis , Neurons/physiology , Spinal Cord/growth & development , Zebrafish/growth & development , Animals , Calcium Signaling , Neural Pathways/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Receptor, Fibroblast Growth Factor, Type 4/physiology , Synapses/physiology , Zebrafish Proteins/physiology
2.
J Orthop Res ; 37(12): 2550-2560, 2019 12.
Article in English | MEDLINE | ID: mdl-31373395

ABSTRACT

Osteoarthritis (OA) is a degenerative joint disease associated with chronic pain and disability in humans and companion animals. The canine species can be subdivided into non-chondrodystrophic (NCD) and chondrodystrophic (CD) dogs, the latter having disproportionally short limbs due to disturbance in endochondral ossification of long bones. This phenotype is associated with retrogene insertions of the fibroblast growth factor 4 (FGF4) gene, resulting in enhanced fibroblast growth factor receptor 3 (FGFR3) signaling. The effect on cartilage is unknown and in experimental studies with dogs, breeds are seemingly employed randomly. The aim of this study was to determine whether CD- and NCD-derived cartilage differs on a structural and biochemical level, and to explore the relationship between FGF4 associated chondrodystrophy and OA. Cartilage explants from CD and NCD dogs were cultured for 21 days. Activation of canonical Wnt signaling was assessed in primary canine chondrocytes. OA and synovitis severity from an experimental OA model were compared between healthy and OA samples from CD and NCD dogs. Release of glycosaminoglycans, DNA content, and cyclooxygenase 2 (COX-2) expression were higher in NCD cartilage explants. Healthy cartilage from NCD dogs displayed higher cartilage degeneration and synovitis scores, which was aggravated by the induction of OA. Dikkopf-3 gene expression was higher in NCD cartilage. No differences in other Wnt pathway read outs were found. To conclude, chondrodystrophy associated with the FGF4 retrogene seems to render CD dogs less susceptible to the development of OA when compared with NCD dogs. These differences should be considered when choosing a canine model to study the pathobiology and new treatment strategies of OA. © 2019 The Authors. Journal of Orthopaedic Research® Published by Wiley Periodicals, Inc. J Orthop Res 37:2550-2560, 2019.


Subject(s)
Disease Models, Animal , Fibroblast Growth Factor 4/genetics , Osteoarthritis/etiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Cartilage, Articular/pathology , Cyclooxygenase 2/analysis , Dogs , Glycosaminoglycans/metabolism , Receptor, Fibroblast Growth Factor, Type 3/physiology , Wnt Signaling Pathway
3.
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
4.
J Neurosci ; 39(23): 4606-4623, 2019 06 05.
Article in English | MEDLINE | ID: mdl-30902874

ABSTRACT

Major depressive disorder is the most common mental illness. Mounting evidence indicates that astrocytes play a crucial role in the pathophysiology of depression; however, the underlying molecular mechanisms remain elusive. Compared with other neuronal cell types, astrocytes are enriched for arachidonic acid metabolism. Herein, we observed brain-region-specific alterations of epoxyeicosatrienoic acid (EET) signaling, which is an arachidonic acid metabolic pathway, in both a mouse model of depression and postmortem samples from patients with depression. The enzymatic activity of soluble epoxide hydrolase (sEH), the key enzyme in EET signaling, was selectively increased in the mPFC of susceptible mice after chronic social defeated stress and was negatively correlated with the social interaction ratio, which is an indicator of depressive-like behavior. The specific deletion of Ephx2 (encode sEH) in adult astrocytes induced resilience to stress, whereas the impaired EET signaling in the mPFC evoked depressive-like behaviors in response to stress. sEH was mainly expressed on lysosomes of astrocytes. Using pharmacological and genetic approaches performed on C57BL/6J background adult male mice, we found that EET signaling modulated astrocytic ATP release in vitro and in vivo Moreover, astrocytic ATP release was required for the antidepressant-like effect of Ephx2 deletion in adult astrocytes. In addition, sEH inhibitors produced rapid antidepressant-like effects in multiple animal models of depression, including chronic social defeated stress and chronic mild stress. Together, our results highlight that EET signaling in astrocytes in the mPFC is essential for behavioral adaptation in response to psychiatric stress.SIGNIFICANCE STATEMENT Astrocytes, the most abundant glial cells of the brain, play a vital role in the pathophysiology of depression. Astrocytes secrete adenosine ATP, which modulates depressive-like behaviors. Notably, astrocytes are enriched for arachidonic acid metabolism. In the present study, we explored the hypothesis that epoxyeicosatrienoic acid signaling, an arachidonic acid metabolic pathway, modulates astrocytic ATP release and the expression of depressive-like behaviors. Our work demonstrated that epoxyeicosatrienoic acid signaling in astrocytes in the mPFC is essential for behavioral homeostatic adaptation in response to stress, and the extent of astrocyte functioning is greater than expected based on earlier reports.


Subject(s)
Astrocytes/metabolism , Depressive Disorder, Major/physiopathology , Eicosanoids/physiology , Prefrontal Cortex/physiology , Adult , Animals , Arachidonic Acids/metabolism , Behavior, Animal/drug effects , Brain Chemistry , Cells, Cultured , Depressive Disorder, Major/genetics , Disease Models, Animal , Double-Blind Method , Eicosanoids/analysis , Epoxide Hydrolases/deficiency , Epoxide Hydrolases/genetics , Epoxide Hydrolases/physiology , Genes, Reporter , Genetic Vectors/administration & dosage , Humans , Lentivirus/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Prefrontal Cortex/chemistry , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/physiology , Signal Transduction , Stress, Psychological/metabolism , Stress, Psychological/psychology , Suicide , Young Adult
5.
J Virol ; 93(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30842331

ABSTRACT

The papillomavirus (PV) E2 protein coordinates viral transcription and genome replication. Following a strategy to identify amino acids in E2 that are posttranslationally modified, we reported that tyrosine kinase fibroblast growth factor receptor 3 (FGFR3) complexes with and phosphorylates E2, which inhibits viral DNA replication. Here, we present several lines of evidence indicating that tyrosine (Y) 138 of HPV-31 E2 is a substrate of FGFR3. The active form of FGFR3 bound to and phosphorylated the region of amino acids (aa) 107 to 175 in HPV-31 E2. The E2 phenylalanine (F) mutant Y138F displayed reduced FGFR3-induced phosphotyrosine. A constitutive kinase-active FGFR3 inhibited wild-type (WT) E2-induced E1-dependent DNA replication, while the 138F mutant retained activity. The tyrosine to glutamic acid (E) mutant Y138E, which can mimic phosphotyrosine, failed to induce transient DNA replication, although it maintained the ability to bind and localize the viral DNA helicase E1 to the viral origin. The bromodomain-containing protein 4 (Brd4) binds to E2 and is necessary for initiation of viral DNA synthesis. Interestingly, the Y138E protein coimmunoprecipitated with full-length Brd4 but was defective for association with its C-terminal domain (CTD). These results imply that the activity of the FGFR3 kinase in the infected epithelial cell restricts the HPV replication program through phosphorylation of E2 at Y138, which interferes with E2 binding to the Brd4 CTD, and that this interaction is required for initiation of viral DNA synthesis.IMPORTANCE Human papillomaviruses (HPVs) are highly infectious pathogens that commonly infect the oropharynx and uterine cervix. The idea that posttranslational modifications of viral proteins coordinates viral genome replication is less explored. We recently discovered that fibroblast growth factor receptor 3 (FGFR3) phosphorylates the viral E2 protein. The current study demonstrates that FGFR3 phosphorylates E2 at tyrosine 138, which inhibits association with the C-terminal peptide of Brd4. This study illustrates a novel regulatory mechanism of virus-host interaction and provides insight into the role of Brd4 in viral replication.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Transcription Factors/metabolism , Virus Replication/physiology , Cell Cycle Proteins/physiology , DNA Helicases/metabolism , DNA Replication , DNA, Viral/metabolism , DNA-Binding Proteins/physiology , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/physiology , Papillomaviridae/metabolism , Papillomaviridae/pathogenicity , Papillomavirus Infections/metabolism , Phosphorylation , Protein Binding , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Fibroblast Growth Factor, Type 3/physiology , Transcription Factors/physiology , Tyrosine/metabolism , Virus Replication/genetics
6.
J Biol Chem ; 294(4): 1142-1151, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30487289

ABSTRACT

CRISPR/Cas9 is a powerful technology widely used for genome editing, with the potential to be used for correcting a wide variety of deleterious disease-causing mutations. However, the technique tends to generate more indels (insertions and deletions) than precise modifications at the target sites, which might not resolve the mutation and could instead exacerbate the initial genetic disruption. We sought to develop an improved protocol for CRISPR/Cas9 that would correct mutations without unintended consequences. As a case study, we focused on achondroplasia, a common genetic form of dwarfism defined by missense mutation in the Fgfr3 gene that results in glycine to arginine substitution at position 374 in mice in fibroblast growth factor receptor 3 (Fgfr3-G374R), which corresponds to G380R in humans. First, we designed a GFP reporter system that can evaluate the cutting efficiency and specificity of single guide RNAs (sgRNAs). Using the sgRNA selected based on our GFP reporter system, we conducted targeted therapy of achondroplasia in mice. We found that we achieved higher frequency of precise correction of the Fgfr3-G374R mutation using Cas9 protein rather than Cas9 mRNA. We further demonstrated that targeting oligos of 100 and 200 nucleotides precisely corrected the mutation at equal efficiency. We showed that our strategy completely suppressed phenotypes of achondroplasia and whole genome sequencing detected no off-target effects. These data indicate that improved protocols can enable the precise CRISPR/Cas9-mediated correction of individual mutations with high fidelity.


Subject(s)
Achondroplasia/therapy , CRISPR-Cas Systems , Gene Targeting , Mutation , Receptor, Fibroblast Growth Factor, Type 3/physiology , Achondroplasia/genetics , Animals , Female , Gene Editing , Male , Mice , Mice, Knockout , Receptor, Fibroblast Growth Factor, Type 3/genetics
7.
Bone ; 111: 71-81, 2018 06.
Article in English | MEDLINE | ID: mdl-29545125

ABSTRACT

Multiple osteochondromas (MO) syndrome is a dominant autosomal bone disorder characterized by the formation of cartilage-capped bony outgrowths that develop at the juxtaposition of the growth plate of endochondral bones. MO has been linked to mutations in either EXT1 or EXT2, two glycosyltransferases required for the synthesis of heparan sulfate (HS). The establishment of mouse mutants demonstrated that a clonal, homozygous loss of Ext1 in a wild type background leads to the development of osteochondromas. Here we investigate mechanisms that might contribute to the variation in the severity of the disease observed in human patients. Our results show that residual amounts of HS are sufficient to prevent the development of osteochondromas strongly supporting that loss of heterozygosity is required for osteochondroma formation. Furthermore, we demonstrate that different signaling pathways affect size and frequency of the osteochondromas thereby modulating the severity of the disease. Reduced Fgfr3 signaling, which regulates proliferation and differentiation of chondrocytes, increases osteochondroma number, while activated Fgfr3 signaling reduces osteochondroma size. Both, activation and reduction of Wnt/ß-catenin signaling decrease osteochondroma size and frequency by interfering with the chondrogenic fate of the mutant cells. Reduced Ihh signaling does not change the development of the osteochondromas, while elevated Ihh signaling increases the cellularity and inhibits chondrocyte differentiation in a subset of osteochondromas and might thus predispose osteochondromas to the transformation into chondrosarcomas.


Subject(s)
Exostoses, Multiple Hereditary/pathology , Hedgehog Proteins/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , beta Catenin/physiology , Animals , Cell Differentiation , Chondrocytes/pathology , Disease Models, Animal , Exostoses, Multiple Hereditary/genetics , Exostoses, Multiple Hereditary/metabolism , Growth Plate/pathology , Hedgehog Proteins/genetics , Heparitin Sulfate/metabolism , Humans , Loss of Heterozygosity , Mice , N-Acetylglucosaminyltransferases/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Signal Transduction , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/genetics
8.
J Orthop Res ; 36(1): 300-308, 2018 01.
Article in English | MEDLINE | ID: mdl-28520086

ABSTRACT

The purpose of this study is to investigate the morphometric changes of the subchondral bone during the development of osteoarthritis (OA) in transgenic mice with achondroplasia (Fgfr3ach ) carrying a heterozygous gain-of-function mutation in Fgfr3. Two OA models (spontaneously developed with age: The aging model, and surgically induced by destabilization of the medial meniscus: The DMM model) were established. Articular cartilage, epiphysis, and metaphysis of the knee joint were histologically and morphometrically compared between wild-type mice, and Fgfr3ach mice in both OA models. Articular cartilage degeneration was scored according to the Osteoarthritis Research Society International (OARSI) scoring system. Several morphometric parameters including bone mineral density (BMD), bone volume/tissue volume (BV/TV), trabecular bone thickness (Tb.Th), and subchondral bone thickness in the medial tibial plateau (MTP) (Sb.Th med) were quantified by micro-computed tomography (CT). In the aging model, although there were no significant differences in the OARSI score between wild-type mice and Fgfr3ach mice, Sb.Th med and Tb.Th in the epiphysis significantly increased in wild-type mice. In the DMM model, the OARSI score of the medial compartment was significantly lower in Fgfr3ach mice than in wild-type mice. BMD, BV/TV, and Tb.Th in the epiphysis increased in wild-type mice and unchanged in Fgfr3ach mice, and the Sb.Th med was significantly larger in wild-type mice after surgery. Subchondral sclerosis, which preceded the cartilage degeneration, was inhibited in Fgfr3ach mice. Activated FGFR3 signaling prevented sclerotic changes of the subchondral bone and subsequent cartilage degeneration. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:300-308, 2018.


Subject(s)
Achondroplasia/pathology , Bone and Bones/pathology , Cartilage, Articular/pathology , Osteoarthritis/pathology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Animals , Disease Models, Animal , Epiphyses/pathology , Male , Mice , Mice, Transgenic , Sclerosis
9.
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
10.
Dev Dyn ; 246(4): 291-309, 2017 04.
Article in English | MEDLINE | ID: mdl-27987249

ABSTRACT

Autosomal dominant mutations in fibroblast growth factor receptor 3 (FGFR3) cause achondroplasia (Ach), the most common form of dwarfism in humans, and related chondrodysplasia syndromes that include hypochondroplasia (Hch), severe achondroplasia with developmental delay and acanthosis nigricans (SADDAN), and thanatophoric dysplasia (TD). FGFR3 is expressed in chondrocytes and mature osteoblasts where it functions to regulate bone growth. Analysis of the mutations in FGFR3 revealed increased signaling through a combination of mechanisms that include stabilization of the receptor, enhanced dimerization, and enhanced tyrosine kinase activity. Paradoxically, increased FGFR3 signaling profoundly suppresses proliferation and maturation of growth plate chondrocytes resulting in decreased growth plate size, reduced trabecular bone volume, and resulting decreased bone elongation. In this review, we discuss the molecular mechanisms that regulate growth plate chondrocytes, the pathogenesis of Ach, and therapeutic approaches that are being evaluated to improve endochondral bone growth in people with Ach and related conditions. Developmental Dynamics 246:291-309, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Achondroplasia , Receptor, Fibroblast Growth Factor, Type 3/genetics , Signal Transduction/physiology , Achondroplasia/etiology , Achondroplasia/pathology , Achondroplasia/therapy , Animals , Chondrocytes/metabolism , Growth Plate/cytology , Growth Plate/metabolism , Growth Plate/ultrastructure , Humans , Receptor, Fibroblast Growth Factor, Type 3/physiology
11.
BJU Int ; 118(5): 681-691, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27271022

ABSTRACT

Molecular analysis has identified subsets of urothelial carcinoma (UC) expressing distinct genetic signatures. Genomic alterations in the oncogenic fibroblast growth factor receptor 3 (FGFR3) pathway are among the most well described in UC and have led to extensive and ongoing investigation of FGFR3-targeted therapies in this disease, although no new drugs have yet been approved. Given the unmet need for effective treatments in advanced and metastatic UC, a better understanding of the known molecular alterations of FGFR3 and of the previous and ongoing clinical investigations of this promising target in UC deserves attention. The objective of the present review is to describe the landscape of alterations and biology of FGFR3 in UC, comprehensively summarize the current state of UC clinical trials of FGFR3 inhibitors, and discuss future therapeutic applications. Using the Pubmed and Clinicaltrials.gov databases, articles describing the spectrum and biological activity of FGFR3 genomic alterations and trials of FGFR3 inhibitors in UC were identified. Search terms included 'FGFR3 genomic alterations' and 'urothelial cancer' or 'bladder cancer'. Genomic alterations, including translocations and activating mutations, are increasingly described in advanced and metastatic UC. The majority of clinical trials have been performed in unselected populations; however, recent studies have reported encouraging preliminary data. We argue that routine use of molecular genomic tumour analysis in UC may inform selection of patients for appropriate trials and we further investigate the potential of FGFR3 as a meaningful clinical target for this difficult disease.


Subject(s)
Carcinoma, Transitional Cell/genetics , Genomics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Urologic Neoplasms/genetics , Clinical Trials as Topic , Forecasting , Humans , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/physiology
12.
Hepatology ; 62(6): 1767-78, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26235436

ABSTRACT

UNLABELLED: Fibroblast growth factor receptors (FGFRs) are frequently up-regulated in subsets of hepatocellular carcinoma (HCC). Here, we provide mechanistic insight that FGFR3 splice variants IIIb and IIIc impact considerably on the malignant phenotype of HCC cells. The occurrence of FGFR3 variants was analyzed in human HCC samples. In hepatoma/hepatocarcinoma cell lines, FGFR3 isoforms were overexpressed by lentiviral constructs or down-modulated by small interfering RNA (siRNA; affecting FGFR3-IIIb and -IIIc) or an adenoviral kinase-dead FGFR3-IIIc construct (kdFGFR3). Elevated levels of FGFR3-IIIb and/or -IIIc were found in 53% of HCC cases. FGFR3-IIIb overexpression occurred significantly more often in primary tumors of large (pT2-4) than of small size (pT1). Furthermore, one or both isoforms were enhanced mostly in cases with early tumor infiltration and/or recurrence at the time of surgery or follow-up examinations. In hepatoma/hepatocarcinoma cells, up-regulated FGFR3-IIIb conferred an enhanced capability for proliferation. Both FGFR3-IIIb and FGFR3-IIIc suppressed apoptotic activity, enhanced clonogenic growth, and induced disintegration of the blood/lymph endothelium. The tumorigenicity of cells in severe combined immunodeficiency mice was augmented to a larger degree by variant IIIb than by IIIc. Conversely, siRNA targeting FGFR3 and kdFGFR3 reduced clonogenicity, anchorage-independent growth, and disintegration of the blood/lymph endothelium in vitro. Furthermore, kdFGFR3 strongly attenuated tumor formation in vivo. CONCLUSIONS: Deregulated FGFR3 variants exhibit specific effects in the malignant progression of HCC cells. Accordingly, blockade of FGFR3-mediated signaling may be a promising therapeutic approach to antagonize growth and malignant behavior of HCC cells.


Subject(s)
Carcinoma, Hepatocellular/etiology , Liver Neoplasms/etiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Animals , Carcinoma, Hepatocellular/drug therapy , Humans , Liver Neoplasms/drug therapy , Mice , Mice, SCID , Protein Isoforms , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Tumor Cells, Cultured , Up-Regulation
13.
Rinsho Shinkeigaku ; 54(12): 1066-8, 2014.
Article in Japanese | MEDLINE | ID: mdl-25519963

ABSTRACT

The precise mechanisms of cortical damage in multiple sclerosis (MS) remain unknown. Microglia, the resident immune cells in the central nervous system (CNS), are involved in the chronic neuroinflammation in MS cortical lesions. Microglia produce various inflammatory cytokines such as IFN-γ and IL-ß, reactive oxygen species, and glutamate. IL-ß secretion is induced by NLRP3. ROS is induced by GM-CSF-producing Th17 cells. Glutamate is released via gap junctions. These molecules exert neurotoxicity. Meanwhile, damaged neurons produce fractalkine and FGF-2, which suppress microglial activation and enhance microglial neuroprotection through anti-inflammatory and anti-oxidant effect. Fractalkine accelerates microglial clearance of neuronal debris via inducing the release of MFG-E8. FGF-2 induces microglial migration through the FGFR3-Wnt-ERK signaling pathway. These molecules suppress microglial neuroinflammation, and enhance neuroprotection, which may give us clues for future therapeutic strategy cortical damage in MS.


Subject(s)
Cerebral Cortex/pathology , Microglia , Multiple Sclerosis/pathology , Anti-Inflammatory Agents , Antigens, Surface/metabolism , Antioxidants/pharmacology , Carrier Proteins/physiology , Cell Movement/drug effects , Chemokine CX3CL1/metabolism , Chemokine CX3CL1/physiology , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/physiology , Gap Junctions/metabolism , Glutamates/metabolism , Glutamates/toxicity , Humans , Interferon-gamma/metabolism , Interferon-gamma/toxicity , Interleukin-1beta/metabolism , Interleukin-1beta/toxicity , MAP Kinase Signaling System , Microglia/metabolism , Microglia/pathology , Milk Proteins/metabolism , Molecular Targeted Therapy , Multiple Sclerosis/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroprotective Agents , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/toxicity , Receptor, Fibroblast Growth Factor, Type 3/physiology , Th17 Cells/physiology , Wnt Signaling Pathway
14.
PLoS One ; 8(2): e57284, 2013.
Article in English | MEDLINE | ID: mdl-23468956

ABSTRACT

Fibroblast growth factor receptors (FGFRs) are activated by mutation and overexpressed in bladder cancers (BCs), and FGFR inhibitors are currently being evaluated in clinical trials in BC patients. However, BC cells display marked heterogeneity in their responses to FGFR inhibitors, and the biological mechanisms underlying this heterogeneity are not well defined. Here we used a novel inhibitor of FGFRs 1-3 and RNAi to determine the effects of inhibiting FGFR1 or FGFR3 in a panel of human BC cell lines. We observed that FGFR1 was expressed in BC cells that also expressed the "mesenchymal" markers ZEB1 and vimentin, whereas FGFR3 expression was restricted to the E-cadherin- and p63-positive "epithelial" subset. Sensitivity to the growth-inhibitory effects of BGJ-398 was also restricted to the "epithelial" BC cells and it correlated directly with FGFR3 mRNA levels but not with the presence of activating FGFR3 mutations. In contrast, BGJ-398 did not strongly inhibit proliferation but did block invasion in the "mesenchymal" BC cells in vitro. Similarly, BGJ-398 did not inhibit primary tumor growth but blocked the production of circulating tumor cells (CTCs) and the formation of lymph node and distant metastases in mice bearing orthotopically implanted "mesenchymal" UM-UC3 cells. Together, our data demonstrate that FGFR1 and FGFR3 have largely non-overlapping roles in regulating invasion/metastasis and proliferation in distinct "mesenchymal" and "epithelial" subsets of human BC cells. The results suggest that the tumor EMT phenotype will be an important determinant of the biological effects of FGFR inhibitors in patients.


Subject(s)
Cell Division/physiology , Neoplasm Metastasis , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Urinary Bladder Neoplasms/pathology , Animals , Base Sequence , Blotting, Western , Cell Division/drug effects , Cell Line, Tumor , DNA Primers , Female , Gene Expression Profiling , Humans , Mice , Mice, Nude , Mutation , RNA Interference , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Reverse Transcriptase Polymerase Chain Reaction
15.
Am J Med Genet A ; 158A(9): 2336-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22888019

ABSTRACT

Achondroplasia and hypochondroplasia are two of the most common forms of skeletal dysplasia. They are both caused by activating mutations in FGFR3 and are inherited in an autosomal dominant manner. Our patient was born to parents with presumed achondroplasia, and found on prenatal testing to have p.G380R and p.N540K FGFR3 mutations. In addition to having typical problems associated with both achondroplasia and hypochondroplasia, our patient had several atypical findings including: abnormal lobulation of the lungs with respiratory insufficiency, C1 stenosis, and hypoglycemia following a Nissen fundoplication. After his reflux and aspiration were treated, the persistence of the tachypnea and increased respiratory effort indicated this was not the primary source of the respiratory distress. Our subsequent hypothesis was that primary restrictive lung disease was the cause of his respiratory distress. A closer examination of his chest circumference did not support this conclusion either. Following his death, an autopsy found the right lung had 2 lobes while the left lung had 3 lobes. A literature review demonstrates that other children with achondroplasia-hypochondroplasia complex have been described with abnormal pulmonary function and infants with thanatophoric dysplasia have similar abnormal pulmonary anatomy. We hypothesize that there may be a primary pulmonary phenotype associated with FGFR3-opathies, unrelated to chest size which leads to the consistent finding of increased respiratory signs and symptoms in these children. Further observation of respiratory status, combined with the macroscopic and microscopic analysis of pulmonary branching anatomy and alveolar structure in this patient population will be important to explore this hypothesis.


Subject(s)
Achondroplasia/pathology , Lung/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Female , Humans , Infant, Newborn , Mutation , Receptor, Fibroblast Growth Factor, Type 3/genetics
16.
Dev Dyn ; 240(11): 2584-96, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22016144

ABSTRACT

Muenke syndrome caused by the FGFR3(P250R) mutation is an autosomal dominant disorder mostly identified with coronal suture synostosis, but it also presents with other craniofacial phenotypes that include mild to moderate midface hypoplasia. The Muenke syndrome mutation is thought to dysregulate intramembranous ossification at the cranial suture without disturbing endochondral bone formation in the skull. We show in this study that knock-in mice harboring the mutation responsible for the Muenke syndrome (FgfR3(P244R)) display postnatal shortening of the cranial base along with synchondrosis growth plate dysfunction characterized by loss of resting, proliferating and hypertrophic chondrocyte zones and decreased Ihh expression. Furthermore, premature conversion of resting chondrocytes along the perichondrium into prehypertrophic chondrocytes leads to perichondrial bony bridge formation, effectively terminating the postnatal growth of the cranial base. Thus, we conclude that the Muenke syndrome mutation disturbs endochondral and perichondrial ossification in the cranial base, explaining the midface hypoplasia in patients.


Subject(s)
Craniosynostoses/genetics , Ossification, Heterotopic/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Skull Base/abnormalities , Amino Acid Substitution/physiology , Animals , Arginine/genetics , Cranial Sutures/abnormalities , Cranial Sutures/diagnostic imaging , Cranial Sutures/metabolism , Cranial Sutures/pathology , Growth Plate/diagnostic imaging , Growth Plate/metabolism , Mice , Mice, Transgenic , Models, Biological , Mutation, Missense/physiology , Osteogenesis/genetics , Phenotype , Proline/genetics , Receptor, Fibroblast Growth Factor, Type 3/physiology , Skull Base/diagnostic imaging , Skull Base/metabolism , X-Ray Microtomography
17.
Dev Dyn ; 240(6): 1586-99, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21491541

ABSTRACT

The fibroblast growth factor receptor 3 (Fgfr3) is expressed in a rostral(low) to caudal(high) gradient in the developing cerebral cortex. Therefore, we hypothesized that Fgfr3 contributes to the correct morphology and connectivity of the caudal cortex. Overall, the forebrain structures appeared normal in Fgfr3(-/-) mice. However, cortical and hippocampal volumes were reduced by 26.7% and 16.3%, respectively. Hypoplasia was particularly evident in the caudo-ventral region of the telencephalon where proliferation was mildly decreased at embryonic day 18.5. Dysplasia of GABAergic neurons in the amygdala and piriform cortex was seen following GAD67 immunohistochemistry. Dye-tracing studies and diffusion magnetic resonance imaging and tractography detected a subtle thalamocortical tract deficit, and significant decreases in the stria terminalis and lateral arms of the anterior commissure. These results indicate the subtle role of Fgfr3 in formation of caudal regions of the telencephalon affecting some brain projections.


Subject(s)
Receptor, Fibroblast Growth Factor, Type 3/physiology , Telencephalon/embryology , Animals , Brain/embryology , Brain/growth & development , Brain/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Down-Regulation , Embryonic Development/genetics , Embryonic Development/physiology , Hippocampus/embryology , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Neural Pathways/metabolism , Neural Pathways/physiology , Neurogenesis/genetics , Neurogenesis/physiology , Organ Size/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Telencephalon/metabolism
18.
Am J Physiol Endocrinol Metab ; 300(3): E508-17, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21139072

ABSTRACT

Uncertainty exists regarding the physiologically relevant fibroblast growth factor (FGF) receptor (FGFR) for FGF23 in the kidney and the precise tubular segments that are targeted by FGF23. Current data suggest that FGF23 targets the FGFR1c-Klotho complex to coordinately regulate phosphate transport and 1,25-dihydroxyvitamin D [1,25(OH)(2)D] production in the proximal tubule. In studies using the Hyp mouse model, which displays FGF23-mediated hypophosphatemia and aberrant vitamin D, deletion of Fgfr3 or Fgfr4 alone failed to correct the Hyp phenotype. To determine whether FGFR1 is sufficient to mediate the renal effects of FGF23, we deleted Fgfr3 and Fgfr4 in Hyp mice, leaving intact the FGFR1 pathway by transferring compound Fgfr3/Fgfr4-null mice on the Hyp background to create wild-type (WT), Hyp, Fgfr3(-/-)/Fgfr4(-/-), and Hyp/Fgfr3(-/-)/Fgfr4(-/-) mice. We found that deletion of Fgfr3 and Fgfr4 in Fgfr3(-/-)/Fgfr4(-/-) and Hyp/Fgfr3(-/-)/Fgfr4(-/-) mice induced an increase in 1,25(OH)(2)D. In Hyp/Fgfr3(-/-)/Fgfr4(-/-) mice, it partially corrected the hypophosphatemia (P(i) = 9.4 ± 0.9, 6.1 ± 0.2, 9.1 ± 0.4, and 8.0 ± 0.5 mg/dl in WT, Hyp, Fgfr3(-/-)/Fgfr4(-/-), and Hyp/Fgfr3(-/-)/Fgfr4(-/-) mice, respectively), increased Na-phosphate cotransporter Napi2a and Napi2c and Klotho mRNA expression in the kidney, and markedly increased serum FGF23 levels (107 ± 20, 3,680 ± 284, 167 ± 22, and 18,492 ± 1,547 pg/ml in WT, Hyp, Fgfr3(-/-)/Fgfr4(-/-), and Hyp/Fgfr3(-/-)/Fgfr4(-/-) mice, respectively), consistent with a compensatory response to the induction of end-organ resistance. Fgfr1 expression was unchanged in Hyp/Fgfr3(-/-)/Fgfr4(-/-) mice and was not sufficient to transduce the full effects of FGF23 in Hyp/Fgfr3(-/-)/Fgfr4(-/-) mice. These studies suggest that FGFR1, FGFR3, and FGFR4 act in concert to mediate FGF23 effects on the kidney and that loss of FGFR function leads to feedback stimulation of Fgf23 expression in bone.


Subject(s)
Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 4/genetics , Absorptiometry, Photon , Animals , Bone and Bones/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/pharmacology , Gene Deletion , Homozygote , Hypophosphatemia/genetics , Hypophosphatemia/metabolism , Immunohistochemistry , Kidney/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Phenotype , Phosphates/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Receptor, Fibroblast Growth Factor, Type 4/physiology , Reverse Transcriptase Polymerase Chain Reaction , Tomography, X-Ray Computed , Vitamin D/metabolism
19.
Clin Calcium ; 20(10): 1490-6, 2010 Oct.
Article in Japanese | MEDLINE | ID: mdl-20890030

ABSTRACT

FGFR3 has been establishing its position in growth plate cartilage after the identification as a responsible gene for achondroplasia. The major pathway of the pathogenesis in achondroplasia is the suppression of PTHrP-PTHR system, which is mainly mediated by ERK activation induced by constitutive active FGFR3. However, intracellular signaling system in FGFR3 is complex and the molecular pathogenesis of achondroplasia and related disorders has not been fully clarified. Especially, recently found human loss-of-function mutations in newly identified syndromes casted novel findings in the relation between phenotype and receptor function. In this review, I summarized recent consensus in the pathogenesis of FGFR3 related chondrodysplasia.


Subject(s)
Achondroplasia/genetics , Mutation , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/physiology , Signal Transduction/physiology , Animals , Cartilage , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/physiology , Growth Plate , Humans , Immunoglobulin D , Mice , Parathyroid Hormone-Related Protein , Receptor, Fibroblast Growth Factor, Type 3/chemistry , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Parathyroid Hormone, Type 1
20.
Anticancer Res ; 30(6): 1921-30, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20651335

ABSTRACT

OBJECTIVE: Ectopically expressed and deregulated fibroblast growth factor receptor 3 (FGFR3) has been observed in many malignant cancer patients, including those with lymphoma. This study investigated whether the therapeutic effect of bortezomib in lymphoma is associated with FGFR3-expression. MATERIALS AND METHODS: Cell proliferation and apoptosis assays were performed in minimal FGFR3 expressing U937 cells and compared to U937 cells overexpressing FGFR3 wild-type, or Y373C or K650E mutant FGFR3. RESULTS: Results from this study suggested the expression of FGFR3 protein is associated with the therapeutic effect of bortezomib. It was observed that bortezomib-induced apoptotic death is correlated with FGFR3 expression. U937 cells overexpression of wild-type FGFR3 demonstrated resistance to bortezomib treatment. U937 cells expressing Y373C mutated FGFR3 showed an almost equal resistance to bortezomib as U937 cells expressing wild-type FGFR3. U937 cells expressing mutated K650E FGFR3 showed more sensitivity to bortezomib than did the parental U937 cells. Furthermore, increased expression of Mcl-1 and decreased expression of NF-kappaB p65 suggested that bortezomib resistance associated with Y373C mutation and wild-type FGFR3 may be partly mediated through Bcl-2 and NF-kappaB signaling. CONCLUSION: Data from this study indicate that mutation status and the expression level of FGFR3 may be associated with bortezomib-related treatment resistance in lymphoma.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Lymphoma/drug therapy , Mutation , Pyrazines/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/genetics , Apoptosis/drug effects , Bortezomib , Drug Resistance, Neoplasm , Humans , Lymphoma/genetics , Myeloid Cell Leukemia Sequence 1 Protein , NF-kappa B/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Receptor, Fibroblast Growth Factor, Type 3/physiology , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/physiology , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...