Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Proc Natl Acad Sci U S A ; 116(10): 4316-4325, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30782830

ABSTRACT

Vertebrate primary cilium is a Hedgehog signaling center but the extent of its involvement in other signaling systems is less well understood. This report delineates a mechanism by which fibroblast growth factor (FGF) controls primary cilia. Employing proteomic approaches to characterize proteins associated with the FGF-receptor, FGFR3, we identified the serine/threonine kinase intestinal cell kinase (ICK) as an FGFR interactor. ICK is involved in ciliogenesis and participates in control of ciliary length. FGF signaling partially abolished ICK's kinase activity, through FGFR-mediated ICK phosphorylation at conserved residue Tyr15, which interfered with optimal ATP binding. Activation of the FGF signaling pathway affected both primary cilia length and function in a manner consistent with cilia effects caused by inhibition of ICK activity. Moreover, knockdown and knockout of ICK rescued the FGF-mediated effect on cilia. We provide conclusive evidence that FGF signaling controls cilia via interaction with ICK.


Subject(s)
Cilia/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , CRISPR-Cas Systems , Fibroblast Growth Factors/metabolism , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Mice , Mice, Knockout , Models, Animal , Molecular Docking Simulation , NIH 3T3 Cells , Phosphorylation , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Proteomics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction
2.
Toxicol Sci ; 168(1): 241-251, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30500929

ABSTRACT

Cylindrospermopsin (CYN) has been recognized as a potent waterborne hepatotoxin with an increasing environmental occurrence. However, CYN effects on the specific populations of hepatic cells involved in liver tissue development, renewal, and regeneration, have not been characterized yet. We used human embryonic stem cells to analyze the hepatic differentiation stage-specific effect of CYN. Our results strongly suggest that CYN might contribute to the development of chronic adverse outcomes by disrupting liver tissue homeostasis in terms of (1) cellular stress and damage induced in the mature differentiated hepatocytes, which was associated with a necrotic cell death and thus possibly also inflammatory responses; (2) selective elimination of HNF4α+ cells from populations of progenitor cells and immature hepatocytes during hepatic differentiation, which could possibly lead to an impaired liver renewal and regeneration; (3) impaired hepatic functions of immature hepatocytes, such as decreased albumin secretion or increased lipid accumulation, which could contribute to the development of liver steatosis; and (4) survival of the immature and AFP-expressing cells with the limited ability to further differentiate, which could represent a tumor-promoting condition.


Subject(s)
Bacterial Toxins/toxicity , Cell Differentiation/drug effects , Hepatocytes/drug effects , Uracil/analogs & derivatives , Albumins/metabolism , Alkaloids , Apoptosis , Cell Survival/drug effects , Cyanobacteria , Cyanobacteria Toxins , Fresh Water , Hepatocyte Nuclear Factor 4/metabolism , Human Embryonic Stem Cells , Humans , Lipid Metabolism , Liver/drug effects , Necrosis , Oxidative Stress/drug effects , Stem Cells , Uracil/toxicity
3.
Sci Rep ; 8(1): 12242, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30115964

ABSTRACT

Yersiniosis belongs to the common foodborne diseases around the world, and frequently manifests as diarrhea that can be treated with probiotics. Colicin FY is an antibacterial agent produced by bacteria and it is capable of specific growth inhibition of Yersinia enterocolitica, the causative agent of gastrointestinal yersiniosis. In this study, recombinant E. coli producing colicin FY were constructed, using both known probiotic strains EcH22 and EcColinfant, and the newly isolated murine strains Ec1127 and Ec1145. All E. coli strains producing colicin FY inhibited growth of pathogenic Y. enterocolitica during co-cultivation in vitro. In dysbiotic mice treated with streptomycin, E. coli strains producing colicin FY inhibited progression of Y. enterocolitica infections. This growth inhibition was not observed in mice with normal gut microflora, likely due to insufficient colonization capacity of E. coli strains and/or due to spatial differences in intestinal niches. Isogenic Y. enterocolitica producing colicin FY was constructed and shown to inhibit pathogenic Y. enterocolitica in mice with normal microflora. Evidence of in vivo antimicrobial activity of colicin FY may have utility in the treatment of Y. enterocolitica infections.


Subject(s)
Colicins/metabolism , Yersinia enterocolitica/physiology , Animals , DNA, Recombinant/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/physiology , Intestines/microbiology , Mice
4.
Biomaterials ; 176: 106-121, 2018 09.
Article in English | MEDLINE | ID: mdl-29879652

ABSTRACT

The blocking of specific protein-protein interactions using nanoparticles is an emerging alternative to small molecule-based therapeutic interventions. However, the nanoparticles designed as "artificial proteins" generally require modification of their surface with (bio)organic molecules and/or polymers to ensure their selectivity and specificity of action. Here, we show that nanosized diamond crystals (nanodiamonds, NDs) without any synthetically installed (bio)organic interface enable the specific and efficient targeting of the family of extracellular signalling molecules known as fibroblast growth factors (FGFs). We found that low nanomolar solutions of detonation NDs with positive ζ-potential strongly associate with multiple FGF ligands present at sub-nanomolar concentrations and effectively neutralize the effects of FGF signalling in cells without interfering with other growth factor systems and serum proteins unrelated to FGFs. We identified an evolutionarily conserved FGF recognition motif, ∼17 amino acids long, that contributes to the selectivity of the ND-FGF interaction. In addition, we inserted this motif into a de novo constructed chimeric protein, which significantly improved its interaction with NDs. We demonstrated that the interaction of NDs, as purely inorganic nanoparticles, with proteins can mitigate pathological FGF signalling and promote the restoration of cartilage growth in a mouse limb explant model. Based on our observations, we foresee that NDs may potentially be applied as nanotherapeutics to neutralize disease-related activities of FGFs in vivo.


Subject(s)
Fibroblast Growth Factors/metabolism , Nanodiamonds/chemistry , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Motifs , Animals , Cartilage/physiology , Cell Line , Cell Proliferation , Cell Survival , Embryo, Mammalian , Humans , Ligands , Mice , Protein Binding , Signal Transduction , Tibia/physiology , Tissue Culture Techniques
5.
Hum Mol Genet ; 27(6): 1093-1105, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29360984

ABSTRACT

Cilia project from almost every cell integrating extracellular cues with signaling pathways. Constitutive activation of FGFR3 signaling produces the skeletal disorders achondroplasia (ACH) and thanatophoric dysplasia (TD), but many of the molecular mechanisms underlying these phenotypes remain unresolved. Here, we report in vivo evidence for significantly shortened primary cilia in ACH and TD cartilage growth plates. Using in vivo and in vitro methodologies, our data demonstrate that transient versus sustained activation of FGF signaling correlated with different cilia consequences. Transient FGF pathway activation elongated cilia, while sustained activity shortened cilia. FGF signaling extended primary cilia via ERK MAP kinase and mTORC2 signaling, but not through mTORC1. Employing a GFP-tagged IFT20 construct to measure intraflagellar (IFT) speed in cilia, we showed that FGF signaling affected IFT velocities, as well as modulating cilia-based Hedgehog signaling. Our data integrate primary cilia into canonical FGF signal transduction and uncover a FGF-cilia pathway that needs consideration when elucidating the mechanisms of physiological and pathological FGFR function, or in the development of FGFR therapeutics.


Subject(s)
Achondroplasia/physiopathology , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Thanatophoric Dysplasia/physiopathology , Achondroplasia/genetics , Animals , Cartilage/metabolism , Chondrocytes/metabolism , Cilia/pathology , Cilia/physiology , Ciliopathies/genetics , Ciliopathies/physiopathology , Fibroblast Growth Factors/metabolism , Growth Plate/metabolism , Humans , Mice , NIH 3T3 Cells , Phenotype , Primary Cell Culture , Receptor, Fibroblast Growth Factor, Type 3/genetics , Signal Transduction/physiology , Thanatophoric Dysplasia/genetics
6.
Biotechnol Bioeng ; 115(4): 850-862, 2018 04.
Article in English | MEDLINE | ID: mdl-29278409

ABSTRACT

Fibroblast growth factors (FGFs) serve numerous regulatory functions in complex organisms, and their corresponding therapeutic potential is of growing interest to academics and industrial researchers alike. However, applications of these proteins are limited due to their low stability. Here we tackle this problem using a generalizable computer-assisted protein engineering strategy to create a unique modified FGF2 with nine mutations displaying unprecedented stability and uncompromised biological function. The data from the characterization of stabilized FGF2 showed a remarkable prediction potential of in silico methods and provided insight into the unfolding mechanism of the protein. The molecule holds a considerable promise for stem cell research and medical or pharmaceutical applications.


Subject(s)
Computer-Aided Design , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Protein Engineering , Protein Stability , Amino Acid Sequence , Animals , Computer Simulation , Directed Molecular Evolution , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 2/chemistry , Humans , Point Mutation , Protein Folding
7.
Cell Signal ; 42: 144-154, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29030113

ABSTRACT

Receptor tyrosine kinases (RTKs) form multiprotein complexes that initiate and propagate intracellular signals and determine the RTK-specific signalling patterns. Unravelling the full complexity of protein interactions within the RTK-associated complexes is essential for understanding of RTK functions, yet it remains an understudied area of cell biology. We describe a comprehensive approach to characterize RTK interactome. A single tag immunoprecipitation and phosphotyrosine protein isolation followed by mass-spectrometry was used to identify proteins interacting with fibroblast growth factor receptor 3 (FGFR3). A total of 32 experiments were carried out in two different cell types and identified 66 proteins out of which only 20 (30.3%) proteins were already known FGFR interactors. Using co-immunoprecipitations, we validated FGFR3 interaction with adapter protein STAM1, transcriptional regulator SHOX2, translation elongation factor eEF1A1, serine/threonine kinases ICK, MAK and CCRK, and inositol phosphatase SHIP2. We show that unappreciated signalling mediators exist for well-studied RTKs, such as FGFR3, and may be identified via proteomic approaches described here. These approaches are easily adaptable to other RTKs, enabling identification of novel signalling mediators for majority of the known human RTKs.


Subject(s)
Gene Expression Regulation , Proteomics/methods , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Signal Transduction/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Expression Profiling , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , NIH 3T3 Cells , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Cyclin-Dependent Kinase-Activating Kinase
8.
Stem Cells Dev ; 26(21): 1521-1527, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28835165

ABSTRACT

Human embryonic stem cells (hESCs) represent a promising tool to study functions of genes during development, to model diseases, and to even develop therapies when combined with gene editing techniques such as CRISPR/CRISPR-associated protein-9 nuclease (Cas9) system. However, the process of disruption of gene expression by generation of null alleles is often inefficient and tedious. To circumvent these limitations, we developed a simple and efficient protocol to permanently downregulate expression of a gene of interest in hESCs using CRISPR/Cas9. We selected p53 for our proof of concept experiments. The methodology is based on series of hESC transfection, which leads to efficient downregulation of p53 expression even in polyclonal population (p53 Low cells), here proven by a loss of regulation of the expression of p53 target gene, microRNA miR-34a. We demonstrate that our approach achieves over 80% efficiency in generating hESC clonal sublines that do not express p53 protein. Importantly, we document by a set of functional experiments that such genetically modified hESCs do retain typical stem cells characteristics. In summary, we provide a simple and robust protocol to efficiently target expression of gene of interest in hESCs that can be useful for laboratories aiming to employ gene editing in their hESC applications/protocols.


Subject(s)
CRISPR-Cas Systems , Embryonic Stem Cells/metabolism , Gene Knockout Techniques/methods , Animals , Cell Line , Cells, Cultured , Down-Regulation , Embryonic Stem Cells/cytology , Humans , Mice , MicroRNAs/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Bone ; 105: 57-66, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28826843

ABSTRACT

Tyrosine kinase inhibitors are being developed for therapy of malignancies caused by oncogenic FGFR signaling but little is known about their effect in congenital chondrodysplasias or craniosynostoses that associate with activating FGFR mutations. Here, we investigated the effects of novel FGFR inhibitor, ARQ 087, in experimental models of aberrant FGFR3 signaling in cartilage. In cultured chondrocytes, ARQ 087 efficiently rescued all major effects of pathological FGFR3 activation, i.e. inhibition of chondrocyte proliferation, loss of extracellular matrix and induction of premature senescence. In ex vivo tibia organ cultures, ARQ 087 restored normal growth plate architecture and eliminated the suppressing FGFR3 effect on chondrocyte hypertrophic differentiation, suggesting that it targets the FGFR3 pathway specifically, i.e. without interference with other pro-growth pathways. Moreover, ARQ 087 inhibited activity of FGFR1 and FGFR2 mutants associated with Pfeiffer, Apert and Beare-Stevenson craniosynostoses, and rescued FGFR-driven excessive osteogenic differentiation in mouse mesenchymal micromass cultures or in ex vivo calvarial organ cultures. Our data warrant further development of ARQ 087 for clinical use in skeletal disorders caused by activating FGFR mutations.


Subject(s)
Aniline Compounds/therapeutic use , Cell Differentiation , Chondrocytes/pathology , Craniosynostoses/drug therapy , Craniosynostoses/pathology , Mutation/genetics , Quinazolines/therapeutic use , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction , Aniline Compounds/pharmacology , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell-Free System , Cellular Senescence/drug effects , Chickens , Chondrocytes/drug effects , Chondrocytes/metabolism , Craniosynostoses/genetics , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblast Growth Factor 2/pharmacology , Limb Buds/pathology , Mice , Organ Culture Techniques , Quinazolines/pharmacology , Rats , Skull/pathology , Tibia/drug effects , Tibia/pathology
10.
Stem Cells ; 35(9): 2050-2059, 2017 09.
Article in English | MEDLINE | ID: mdl-28631381

ABSTRACT

Human pluripotent stem cells (hPSC) require signaling provided by fibroblast growth factor (FGF) receptors. This can be initiated by the recombinant FGF2 ligand supplied exogenously, but hPSC further support their niche by secretion of endogenous FGF2. In this study, we describe a role of tyrosine kinase expressed in hepatocellular carcinoma (TEC) kinase in this process. We show that TEC-mediated FGF2 secretion is essential for hPSC self-renewal, and its lack mediates specific differentiation. Following both short hairpin RNA- and small interfering RNA-mediated TEC knockdown, hPSC secretes less FGF2. This impairs hPSC proliferation that can be rescued by increasing amounts of recombinant FGF2. TEC downregulation further leads to a lower expression of the pluripotency markers, an improved priming towards neuroectodermal lineage, and a failure to develop cardiac mesoderm. Our data thus demonstrate that TEC is yet another regulator of FGF2-mediated hPSC pluripotency and differentiation. Stem Cells 2017;35:2050-2059.


Subject(s)
Cell Lineage , Fibroblast Growth Factor 2/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/enzymology , Protein-Tyrosine Kinases/metabolism , Biomarkers/metabolism , Cell Line , Cell Lineage/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Humans , Recombinant Proteins/pharmacology
11.
Elife ; 62017 02 15.
Article in English | MEDLINE | ID: mdl-28199182

ABSTRACT

In-cell profiling enables the evaluation of receptor tyrosine activity in a complex environment of regulatory networks that affect signal initiation, propagation and feedback. We used FGF-receptor signaling to identify EGR1 as a locus that strongly responds to the activation of a majority of the recognized protein kinase oncogenes, including 30 receptor tyrosine kinases and 154 of their disease-associated mutants. The EGR1 promoter was engineered to enhance trans-activation capacity and optimized for simple screening assays with luciferase or fluorescent reporters. The efficacy of the developed, fully synthetic reporters was demonstrated by the identification of novel targets for two clinically used tyrosine kinase inhibitors, nilotinib and osimertinib. A universal reporter system for in-cell protein kinase profiling will facilitate repurposing of existing anti-cancer drugs and identification of novel inhibitors in high-throughput screening studies.


Subject(s)
Cytological Techniques/methods , Oncogene Proteins/analysis , Protein Kinases/analysis , Animals , Cell Line , Humans , Intravital Microscopy , Mice , Optical Imaging
12.
Oncotarget ; 8(65): 109319-109331, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29312610

ABSTRACT

Many tyrosine kinase inhibitors (TKIs) have failed to reach human use due to insufficient activity in clinical trials. However, the failed TKIs may still benefit patients if their other kinase targets are identified by providing treatment focused on syndromes driven by these kinases. Here, we searched for novel targets of AZD1480, an inhibitor of JAK2 kinase that recently failed phase two cancer clinical trials due to a lack of activity. Twenty seven human receptor tyrosine kinases (RTKs) and 153 of their disease-associated mutants were in-cell profiled for activity in the presence of AZD1480 using a newly developed RTK plasmid library. We demonstrate that AZD1480 inhibits ALK, LTK, FGFR1-3, RET and TRKA-C kinases and uncover a physical basis of this specificity. The RTK activity profiling described here facilitates inhibitor repurposing by enabling rapid and efficient identification of novel TKI targets in cells.

13.
Stem Cells Dev ; 26(5): 328-340, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27863459

ABSTRACT

HMGB1 and HMGB2 proteins have been implicated in numerous cellular processes, including proliferation, differentiation, apoptosis, and tumor growth. It is unknown whether they are involved in regulating the typical functions of pluripotent human embryonic stem cells (hESCs) and/or those of the differentiated derivatives of hESCs. Using inducible, stably transfected hESCs capable of shRNA-mediated knockdown of HMGB1 and HMGB2, we provide evidence that downregulation of HMGB1 and/or HMGB2 in undifferentiated hESCs does not affect the stemness of cells and induces only minor changes to the proliferation rate, cell-cycle profile, and apoptosis. After differentiation is induced, however, the downregulation of those proteins has important effects on proliferation, apoptosis, telomerase activity, and the efficiency of differentiation toward the neuroectodermal lineage. Furthermore, those processes are affected only when one, but not both, of the two proteins is downregulated; the knockdown of both HMGB1 and HMGB2 results in a normal phenotype. Those results advance our knowledge of regulation of hESC and human neuroectodermal cell differentiation and illustrate the distinct roles of HMGB1 and HMGB2 during early human development.


Subject(s)
Cell Differentiation , HMGB1 Protein/metabolism , HMGB2 Protein/metabolism , Histones/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Apoptosis/genetics , Cell Cycle/genetics , Cell Line , Cell Lineage/genetics , Cell Proliferation/genetics , Cell Self Renewal/genetics , Cell Shape/genetics , Down-Regulation/genetics , Humans , Neural Plate/cytology , Telomerase/metabolism , Transfection
14.
Front Physiol ; 7: 378, 2016.
Article in English | MEDLINE | ID: mdl-27630576

ABSTRACT

MORN5 (MORN repeat containing 5) is encoded by a locus positioned on chromosome 17 in the chicken genome. The MORN motif is found in multiple copies in several proteins including junctophilins or phosphatidylinositol phosphate kinase family and the MORN proteins themselves are found across the animal and plant kingdoms. MORN5 protein has a characteristic punctate pattern in the cytoplasm in immunofluorescence imaging. Previously, MORN5 was found among differentially expressed genes in a microarray profiling experiment of the chicken embryo head. Here, we provided in situ hybridization to analyse, in detail, the MORN5 expression in chick craniofacial structures. The expression of MORN5 was first observed at stage HH17-18 (E2.5). MORN5 expression gradually appeared on either side of the primitive oral cavity, within the maxillary region. At stage HH20 (E3), prominent expression was localized in the mandibular prominences lateral to the midline. From stage HH20 up to HH29 (E6), there was strong expression in restricted regions of the maxillary and mandibular prominences. The frontonasal mass (in the midline of the face) expressed MORN5, starting at HH27 (E5). The expression was concentrated in the corners or globular processes, which will ultimately fuse with the cranial edges of the maxillary prominences. MORN5 expression was maintained in the fusion zone up to stage HH29. In sections MORN5 expression was localized preferentially in the mesenchyme. Previously, we examined signals that regulate MORN5 expression in the face based on a previous microarray study. Here, we validated the array results with in situ hybridization and QPCR. MORN5 was downregulated 24 h after Noggin and/or RA treatment. We also determined that BMP pathway genes are downstream of MORN5 following siRNA knockdown. Based on these results, we conclude that MORN5 is both regulated by and required for BMP signaling. The restricted expression of MORN5 in the lip fusion zone shown here supports the human genetic data in which MORN5 variants were associated with increased risk of non-syndromic cleft lip with or without cleft palate.

15.
J Proteome Res ; 15(10): 3841-3855, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27615514

ABSTRACT

The fibroblast growth factor receptors (FGFRs) are important oncogenes promoting tumor progression in many types of cancer, such as breast, bladder, and lung cancer as well as multiple myeloma and rhabdomyosarcoma. However, little is known about how these receptors are internalized and down-regulated in cells. We have here applied proximity biotin labeling to identify proteins involved in FGFR4 signaling and trafficking. For this purpose we fused a mutated biotin ligase, BirA*, to the C-terminal tail of FGFR4 (FGFR4-BirA*) and the fusion protein was stably expressed in U2OS cells. Upon addition of biotin to these cells, proteins in proximity to the FGFR4-BirA* fusion protein became biotinylated and could be isolated and identified by quantitative mass spectrometry. We identified in total 291 proteins, including 80 proteins that were enriched in samples where the receptor was activated by the ligand (FGF1), among them several proteins previously found to be involved in FGFR signaling (e.g., FRS2, PLCγ, RSK2 and NCK2). Interestingly, many of the identified proteins were implicated in endosomal transport, and by precise annotation we were able to trace the intracellular pathways of activated FGFR4. Validating the data by confocal and three-dimensional structured illumination microscopy analysis, we concluded that FGFR4 uses clathrin-mediated endocytosis for internalization and is further sorted from early endosomes to the recycling compartment and the trans-Golgi network. Depletion of cells for clathrin heavy chain led to accumulation of FGFR4 at the cell surface and increased levels of active FGFR4 and PLCγ, while AKT and ERK signaling was diminished, demonstrating that functional clathrin-mediated endocytosis is required for proper FGFR4 signaling. Thus, this study reveals proteins and pathways involved in FGFR4 transport and signaling that provide possible targets and opportunities for therapeutic intervention in FGFR4 aberrant cancer.


Subject(s)
Endosomes/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Biotinylation , Cell Line, Tumor , Clathrin/metabolism , Endocytosis , Humans , Microscopy/methods , Protein Transport , Signal Transduction , Staining and Labeling , trans-Golgi Network/metabolism
16.
Hum Mol Genet ; 25(18): 3998-4011, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27466187

ABSTRACT

The short rib polydactyly syndromes (SRPS) are a group of recessively inherited, perinatal-lethal skeletal disorders primarily characterized by short ribs, shortened long bones, varying types of polydactyly and concomitant visceral abnormalities. Mutations in several genes affecting cilia function cause SRPS, revealing a role for cilia function in skeletal development. To identify additional SRPS genes and discover novel ciliary molecules required for normal skeletogenesis, we performed exome sequencing in a cohort of patients and identified homozygosity for a missense mutation, p.E80K, in Intestinal Cell Kinase, ICK, in one SRPS family. The p.E80K mutation abolished serine/threonine kinase activity, resulting in altered ICK subcellular and ciliary localization, increased cilia length, aberrant cartilage growth plate structure, defective Hedgehog and altered ERK signalling. These data identify ICK as an SRPS-associated gene and reveal that abnormalities in signalling pathways contribute to defective skeletogenesis.


Subject(s)
Abnormalities, Multiple/genetics , Hedgehog Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Short Rib-Polydactyly Syndrome/genetics , Skeleton/growth & development , Abnormalities, Multiple/physiopathology , Cilia/genetics , Cilia/pathology , Exome/genetics , Female , Humans , Infant , MAP Kinase Signaling System , Pedigree , Pregnancy , Sequence Analysis, DNA , Short Rib-Polydactyly Syndrome/pathology , Signal Transduction , Skeleton/abnormalities
SELECTION OF CITATIONS
SEARCH DETAIL
...