Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 575
Filtrar
1.
Endocrinology ; 163(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34977930

RESUMO

Luminal breast cancer (BrCa) has a favorable prognosis compared with other tumor subtypes. However, with time, tumors may evolve and lead to disease progression; thus, there is a great interest in unraveling the mechanisms that drive tumor metastasis and endocrine resistance. In this review, we focus on one of the many pathways that have been involved in tumor progression, the fibroblast growth factor/fibroblast growth factor receptor (FGFR) axis. We emphasize in data obtained from in vivo experimental models that we believe that in luminal BrCa, tumor growth relies in a crosstalk with the stromal tissue. We revisited the studies that illustrate the interaction between hormone receptors and FGFR. We also highlight the most frequent alterations found in BrCa cell lines and provide a short review on the trials that use FGFR inhibitors in combination with endocrine therapies. Analysis of these data suggests there are many players involved in this pathway that might be also targeted to decrease FGF signaling, in addition to specific FGFR inhibitors that may be exploited to increase their efficacy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Fatores de Crescimento de Fibroblastos/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Receptores de Esteroides/fisiologia , Transdução de Sinais/fisiologia , Animais , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/química , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Receptor alfa de Estrogênio/análise , Feminino , Fatores de Crescimento de Fibroblastos/genética , Amplificação de Genes , Humanos , Camundongos , Mutação , Receptor Cross-Talk/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/genética
2.
Theranostics ; 11(20): 10125-10147, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34815808

RESUMO

Background: Fibroblast growth factor receptors (FGFRs) are key targets for nerve regeneration and repair. The therapeutic effect of exogenous recombinant FGFs in vivo is limited due to their high molecular weight. Small peptides with low molecular weight, easy diffusion, low immunogenicity, and nontoxic metabolite formation are potential candidates. The present study aimed to develop a novel low-molecular-weight peptide agonist of FGFR to promote nerve injury repair. Methods: Phage display technology was employed to screen peptide ligands targeting FGFR2. The peptide ligand affinity for FGFRs was detected by isothermal titration calorimetry. Structural biology-based computer virtual analysis was used to characterize the interaction between the peptide ligand and FGFR2. The peptide ligand effect on axon growth, regeneration, and behavioral recovery of sensory neurons was determined in the primary culture of sensory neurons and dorsal root ganglia (DRG) explants in vitro and a rat spinal dorsal root injury (DRI) model in vivo. The peptide ligand binding to other membrane receptors was characterized by surface plasmon resonance (SPR) and liquid chromatography-mass spectrometry (LC-MS)/MS. Intracellular signaling pathways primarily affected by the peptide ligand were characterized by phosphoproteomics, and related pathways were verified using specific inhibitors. Results: We identified a novel FGFR-targeting small peptide, CH02, with seven amino acid residues. CH02 activated FGFR signaling through high-affinity binding with the extracellular segment of FGFRs and also had an affinity for several receptor tyrosine kinase (RTK) family members, including VEGFR2. In sensory neurons cultured in vitro, CH02 maintained the survival of neurons and promoted axon growth. Simultaneously, CH02 robustly enhanced nerve regeneration and sensory-motor behavioral recovery after DRI in rats. CH02-induced activation of FGFR signaling promoted nerve regeneration primarily via AKT and ERK signaling downstream of FGFRs. Activation of mTOR downstream of AKT signaling augmented axon growth potential in response to CH02. Conclusion: Our study revealed the significant therapeutic effect of CH02 on strengthening nerve regeneration and suggested a strategy for treating peripheral and central nervous system injuries.


Assuntos
Peptídeos/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Raízes Nervosas Espinhais/efeitos dos fármacos , Animais , Axônios/metabolismo , Células Cultivadas , Lesões por Esmagamento/tratamento farmacológico , Lesões por Esmagamento/metabolismo , Gânglios Espinais/metabolismo , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Ligantes , Masculino , Simulação de Acoplamento Molecular , Regeneração Nervosa/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Raízes Nervosas Espinhais/lesões , Serina-Treonina Quinases TOR/metabolismo
3.
Life Sci Alliance ; 4(11)2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34548382

RESUMO

FGFs and their high-affinity receptors (FGFRs) play key roles in development, tissue repair, and disease. Because FGFRs bind overlapping sets of ligands, their individual functions cannot be determined using ligand stimulation. Here, we generated a light-activated FGFR2 variant (OptoR2) to selectively activate signaling by the major FGFR in keratinocytes. Illumination of OptoR2-expressing HEK 293T cells activated FGFR signaling with remarkable temporal precision and promoted cell migration and proliferation. In murine and human keratinocytes, OptoR2 activation rapidly induced the classical FGFR signaling pathways and expression of FGF target genes. Surprisingly, multi-level counter-regulation occurred in keratinocytes in vitro and in transgenic mice in vivo, including OptoR2 down-regulation and loss of responsiveness to light activation. These results demonstrate unexpected cell type-specific limitations of optogenetic FGFRs in long-term in vitro and in vivo settings and highlight the complex consequences of transferring optogenetic cell signaling tools into their relevant cellular contexts.


Assuntos
Queratinócitos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/fisiologia , Células HEK293 , Humanos , Queratinócitos/fisiologia , Ligantes , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais
4.
Osteoarthritis Cartilage ; 29(10): 1389-1398, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34284112

RESUMO

Osteoarthritis (OA) is a major health problem worldwide that affects the joints and causes severe disability. It is characterized by pain and low-grade inflammation. However, the exact pathogenesis remains unknown and the therapeutic options are limited. In OA articular chondrocytes undergo a phenotypic transition becoming hypertrophic, which leads to cartilage damage, aggravating the disease. Therefore, a therapeutic agent inhibiting hypertrophy would be a promising disease-modifying drug. The therapeutic use of tyrosine kinase inhibitors has been mainly focused on oncology, but the Food and Drug Administration (FDA) approval of the Janus kinase inhibitor Tofacitinib in Rheumatoid Arthritis has broadened the applicability of these compounds to other diseases. Interestingly, tyrosine kinases have been associated with chondrocyte hypertrophy. In this review, we discuss the experimental evidence that implicates specific tyrosine kinases in signaling pathways promoting chondrocyte hypertrophy, highlighting their potential as therapeutic targets for OA.


Assuntos
Condrócitos/patologia , Osteoartrite/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptores com Domínio Discoidina/fisiologia , Receptores ErbB/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Humanos , Hipertrofia/tratamento farmacológico , Janus Quinase 2/fisiologia , Osteoartrite/fisiopatologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/fisiologia , Receptor IGF Tipo 1/fisiologia , Receptor trkA/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais
5.
Physiol Rev ; 101(2): 569-610, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32730114

RESUMO

Fibroblast growth factors (FGFs) are a family of proteins possessing paracrine, autocrine, or endocrine functions in a variety of biological processes, including embryonic development, angiogenesis, tissue homeostasis, wound repair, and cancer. Canonical FGFs bind and activate tyrosine kinase FGF receptors (FGFRs), triggering intracellular signaling cascades that mediate their biological activity. Experimental evidence indicates that FGFs play a complex role in the physiopathology of the prostate gland that ranges from essential functions during embryonic development to modulation of neoplastic transformation. The use of ligand- and receptor-deleted mouse models has highlighted the requirement for FGF signaling in the normal development of the prostate gland. In adult prostate, the maintenance of a functional FGF/FGFR signaling axis is critical for organ homeostasis and function, as its disruption leads to prostate hyperplasia and may contribute to cancer progression and metastatic dissemination. Dissection of the molecular landscape modulated by the FGF family will facilitate ongoing translational efforts directed toward prostate cancer therapy.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Próstata/fisiologia , Próstata/fisiopatologia , Doenças Prostáticas/fisiopatologia , Neoplasias da Próstata/fisiopatologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Animais , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Masculino , Próstata/crescimento & desenvolvimento
6.
Curr Drug Targets ; 22(2): 214-240, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33045958

RESUMO

BACKGROUND: The fibroblast growth factor (FGF) family is comprised of 23 highly regulated monomeric proteins that regulate a plethora of developmental and pathophysiological processes, including tissue repair, wound healing, angiogenesis, and embryonic development. Binding of FGF to fibroblast growth factor receptor (FGFR), a tyrosine kinase receptor, is facilitated by a glycosaminoglycan, heparin. Activated FGFRs phosphorylate the tyrosine kinase residues that mediate induction of downstream signaling pathways, such as RAS-MAPK, PI3K-AKT, PLCγ, and STAT. Dysregulation of the FGF/FGFR signaling occurs frequently in cancer due to gene amplification, FGF activating mutations, chromosomal rearrangements, integration, and oncogenic fusions. Aberrant FGFR signaling also affects organogenesis, embryonic development, tissue homeostasis, and has been associated with cell proliferation, angiogenesis, cancer, and other pathophysiological changes. OBJECTIVE: This comprehensive review will discuss the biology, chemistry, and functions of FGFs, and its current applications toward wound healing, diabetes, repair and regeneration of tissues, and fatty liver diseases. In addition, specific aberrations in FGFR signaling and drugs that target FGFR and aid in mitigating various disorders, such as cancer, are also discussed in detail. CONCLUSION: Inhibitors of FGFR signaling are promising drugs in the treatment of several types of cancers. The clinical benefits of FGF/FGFR targeting therapies are impeded due to the activation of other RTK signaling mechanisms or due to the mutations that abolish the drug inhibitory activity on FGFR. Thus, the development of drugs with a different mechanism of action for FGF/FGFR targeting therapies is the recent focus of several preclinical and clinical studies.


Assuntos
Fatores de Crescimento de Fibroblastos , Neoplasias , Receptores de Fatores de Crescimento de Fibroblastos , Transdução de Sinais , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/fisiologia , Humanos , Neoplasias/tratamento farmacológico , Neovascularização Patológica , Fosfatidilinositol 3-Quinases , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/efeitos dos fármacos
7.
Nat Commun ; 11(1): 4491, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32901033

RESUMO

The functionality of the nervous system requires transmission of information along axons with high speed and precision. Conductance velocity depends on axonal diameter whereas signaling precision requires a block of electrical crosstalk between axons, known as ephaptic coupling. Here, we use the peripheral nervous system of Drosophila larvae to determine how glia regulates axonal properties. We show that wrapping glial differentiation depends on gap junctions and FGF-signaling. Abnormal glial differentiation affects axonal diameter and conductance velocity and causes mild behavioral phenotypes that can be rescued by a sphingosine-rich diet. Ablation of wrapping glia does not further impair axonal diameter and conductance velocity but causes a prominent locomotion phenotype that cannot be rescued by sphingosine. Moreover, optogenetically evoked locomotor patterns do not depend on conductance speed but require the presence of wrapping glial processes. In conclusion, our data indicate that wrapping glia modulates both speed and precision of neuronal signaling.


Assuntos
Drosophila melanogaster/fisiologia , Animais , Animais Geneticamente Modificados , Axônios/fisiologia , Diferenciação Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Larva/citologia , Larva/fisiologia , Locomoção/fisiologia , Modelos Neurológicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Neuroglia/citologia , Neuroglia/fisiologia , Optogenética , Sistema Nervoso Periférico/citologia , Sistema Nervoso Periférico/fisiologia , Fenótipo , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais
8.
Dev Genes Evol ; 230(3): 227-238, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32198667

RESUMO

Across the Bilateria, FGF/FGFR signaling is critical for normal development, and in both Drosophila and vertebrates, docking proteins are required to connect activated FGFRs with downstream pathways. While vertebrates use Frs2 to dock FGFR to the RAS/MAPK or PI3K pathways, the unrelated protein, downstream of FGFR (Dof/stumps/heartbroken), fulfills the corresponding function in Drosophila. To better understand the evolution of the signaling pathway downstream of FGFR, the available sequence databases were screened to identify Frs2, Dof, and other key pathway components in phyla that diverged early in animal evolution. While Frs2 homologues were detected only in members of the Bilateria, canonical Dof sequences (containing Dof, ankyrin, and SH2/SH3 domains) were present in cnidarians as well as bilaterians (but not in other animals or holozoans), correlating with the appearance of FGFR. Although these data suggested that Dof coupling might be ancestral, gene expression analysis in the cnidarian Hydra revealed that Dof is not upregulated in the zone of strong FGFRa and FGFRb expression at the bud base, where FGFR signaling controls detachment. In contrast, transcripts encoding other, known elements of FGFR signaling in Bilateria, namely the FGFR adaptors Grb2 and Crkl, which are acting downstream of Dof (and Frs2), as well as the guanyl nucleotide exchange factor Sos, and the tyrosine phosphatase Csw/Shp2, were strongly upregulated at the bud base. Our expression analysis, thus, identified transcriptional upregulation of known elements of FGFR signaling at the Hydra bud base indicating a highly conserved toolkit. Lack of transcriptional Dof upregulation raises the interesting question, whether Hydra FGFR signaling requires either of the docking proteins known from Bilateria.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Evolução Biológica , Hydra/genética , Hydra/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Animais , Cnidários/genética , Cnidários/metabolismo , Proteína Adaptadora GRB2/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Filogenia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Transdução de Sinais , Proteínas Son Of Sevenless/fisiologia
9.
Biol Cell ; 112(5): 127-139, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32027762

RESUMO

BACKGROUND INFORMATION: The fibroblast growth factor (FGF) signalling system of vertebrates is complex. In common with other vertebrates, secreted FGF ligands of the amphibian Xenopus signal through a family of four FGF receptor tyrosine kinases (fgfr1, 2, 3 and 4). A wealth of previous studies has demonstrated important roles for FGF signalling in regulating gene expression during cell lineage specification in amphibian development. In particular, FGFs have well-established roles in regulating mesoderm formation, neural induction and patterning of the anteroposterior axis. However, relatively little is known regarding the role of individual FGFRs in regulating FGF-dependent processes in amphibian development. In this study we make use of synthetic drug inducible versions of Xenopus Fgfr1, 2 and 4 (iFgfr1, 2 and 4) to undertake a comparative analysis of their activities in the tissues of the developing embryo. RESULTS: We find that Xenopus Fgfr1 and 2 have very similar activities. Both Fgfr1 and Fgfr2 are potent activators of MAP kinase ERK signalling, and when activated in the embryo during gastrula stages regulate similar cohorts of transcriptional targets. In contrast, Fgfr4 signalling in naïve ectoderm and neuralised ectoderm activates ERK signalling only weakly compared to Fgfr1/2. Furthermore, our analyses indicate that in Xenopus neural tissue the Fgfr4 regulated transcriptome is very different from that of Fgfr1. CONCLUSION AND SIGNIFICANCE: We conclude that signalling downstream of Fgfr1 and 2 regulates similar processes in amphibian development. Interestingly, many of the previously identified canonical transcriptional targets of FGF regulation associated with germ layer specification and patterning are regulated by Fgfr1/Fgfr2 signalling. In contrast, the downstream consequences of Fgfr4 signalling are different, although roles for Fgfr4 signalling in lineage specification and anteroposterior patterning are also indicated.


Assuntos
Padronização Corporal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/fisiologia , Xenopus/embriologia , Animais
10.
J Pharm Pharmacol ; 71(9): 1412-1420, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31282010

RESUMO

OBJECTIVES: The basic fibroblast growth factor (bFGF)/fibroblast growth factor receptor (FGFR) signal transductional pathway plays an important role not only in tumour, but also in tumour stem cells. Thus, this study was designed to investigate the effects of bFGF signalling on cancer stem cells of lung cancer. METHODS: We blocked bFGF/FGFR signalling in cisplatin (DDP) selected A549 by knocking down bFGF via RNA interference, and subsequently, the stem cell marker of OCT-4 was determined, and cell proliferation, clone formation, invasiveness, apoptosis and drug resistance abilities of DDP selected A549 cells were investigated. KEY FINDINGS: The expressions of bFGF and OCT-4 in DDP selected A549 were higher than that of A549 cells. The findings suggested blocking of bFGF/FGFR signalling resulted in downregulation of bFGF, reduction in cell proliferation, clone formation, invasion and drug resistance abilities, and increase in cell apoptosis. Furthermore, our results also revealed OCT-4 was reduced after bFGF signalling blocking. CONCLUSIONS: In conclusion, our study suggested that bFGF/FGFR signalling plays an important role in maintaining lung cancer stem cell characteristics and regulating expression of cancer stem cell marker of OCT-4.


Assuntos
Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Células A549 , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Regulação para Baixo/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Fator 2 de Crescimento de Fibroblastos/genética , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/patologia , Paclitaxel/farmacologia , RNA Interferente Pequeno , Transdução de Sinais/efeitos dos fármacos
11.
Cells ; 8(6)2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31216761

RESUMO

Fibroblast growth factor receptors (FGFRs) are a family of receptor tyrosine kinases expressed on the cell membrane that play crucial roles in both developmental and adult cells. Dysregulation of FGFRs has been implicated in a wide variety of cancers, such as urothelial carcinoma, hepatocellular carcinoma, ovarian cancer and lung adenocarcinoma. Due to their functional importance, FGFRs have been considered as promising drug targets for the therapy of various cancers. Multiple small molecule inhibitors targeting this family of kinases have been developed, and some of them are in clinical trials. Furthermore, the pan-FGFR inhibitor erdafitinib (JNJ-42756493) has recently been approved by the U.S. Food and Drug Administration (FDA) for the treatment of metastatic or unresectable urothelial carcinoma (mUC). This review summarizes the structure of FGFR, especially its kinase domain, and the development of small molecule FGFR inhibitors.


Assuntos
Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/fisiologia , Humanos , Inibidores de Proteínas Quinases/química , Pirazóis , Quinoxalinas , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
Cells ; 8(5)2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31091809

RESUMO

Fibroblast growth factors (FGFs) and their receptors (FGFRs) constitute signaling circuits that transmit signals across the plasma membrane, regulating pivotal cellular processes like differentiation, migration, proliferation, and apoptosis. The malfunction of FGFs/FGFRs signaling axis is observed in numerous developmental and metabolic disorders, and in various tumors. The large diversity of FGFs/FGFRs functions is attributed to a great complexity in the regulation of FGFs/FGFRs-dependent signaling cascades. The function of FGFRs is modulated at several levels, including gene expression, alternative splicing, posttranslational modifications, and protein trafficking. One of the emerging ways to adjust FGFRs activity is through formation of complexes with other integral proteins of the cell membrane. These proteins may act as coreceptors, modulating binding of FGFs to FGFRs and defining specificity of elicited cellular response. FGFRs may interact with other cell surface receptors, like G-protein-coupled receptors (GPCRs) or receptor tyrosine kinases (RTKs). The cross-talk between various receptors modulates the strength and specificity of intracellular signaling and cell fate. At the cell surface FGFRs can assemble into large complexes involving various cell adhesion molecules (CAMs). The interplay between FGFRs and CAMs affects cell-cell interaction and motility and is especially important for development of the central nervous system. This review summarizes current stage of knowledge about the regulation of FGFRs by the plasma membrane-embedded partner proteins and highlights the importance of FGFRs-containing membrane complexes in pathological conditions, including cancer.


Assuntos
Moléculas de Adesão Celular/metabolismo , Fatores de Crescimento de Fibroblastos/fisiologia , Receptores de Superfície Celular/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Animais , Linhagem Celular , Movimento Celular/fisiologia , Sistema Nervoso Central/metabolismo , Humanos , Neoplasias/metabolismo , Transdução de Sinais
13.
Curr Osteoporos Rep ; 17(3): 138-146, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30982184

RESUMO

PURPOSE OF REVIEW: Fibroblast growth factor receptor (FGFR) signaling regulates proliferation and differentiation during development and homeostasis. While membrane-bound FGFRs play a central role in these processes, the function of nuclear FGFRs is also critical. Here, we highlight mechanisms for nuclear FGFR translocation and the effects of nuclear FGFRs on skeletal development and disease. RECENT FINDINGS: Full-length FGFRs, internalized by endocytosis, enter the nucleus through ß-importin-dependent mechanisms that recognize the nuclear localization signal within FGFs. Alternatively, soluble FGFR intracellular fragments undergo nuclear translocation following their proteolytic release from the membrane. FGFRs enter the nucleus during the cellular transition between proliferation and differentiation. Once nuclear, FGFRs interact with chromatin remodelers to alter the epigenetic state and transcription of their target genes. Dysregulation of nuclear FGFR is linked to the etiology of congenital skeletal disorders and neoplastic transformation. Revealing the activities of nuclear FGFR will advance our understanding of 20 congenital skeletal disorders caused by FGFR mutations, as well as FGFR-related cancers.


Assuntos
Doenças Ósseas/etiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Osteogênese/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Diferenciação Celular , Humanos , Transdução de Sinais
14.
Dev Dyn ; 248(3): 233-246, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30620790

RESUMO

BACKGROUND: Bent bone dysplasia syndrome (BBDS), a congenital skeletal disorder caused by dominant mutations in fibroblast growth factor receptor 2 (FGFR2), is characterized by bowed long bones within the limbs. We previously showed that the FGFR2 mutations in BBDS enhance nuclear and nucleolar localization of the receptor; however, exactly how shifts in subcellular distribution of FGFR2 affect limb development remained unknown. RESULTS: Targeted expression of the BBDS mutations in the lateral plate mesoderm of the developing chick induced angulated hindlimbs, a hallmark feature of the disease. Whole-mount analysis of the underlying skeleton revealed bent long bones with shortened bone collars and, in severe cases, dysmorphic epiphyses. Epiphyseal changes were also correlated with joint dislocations and contractures. Histological analysis revealed that bent long bones and joint defects were closely associated with irregularities in skeletal muscle patterning and tendon-to-bone attachment. The spectrum of limb phenotypes induced by the BBDS mutations were recapitulated by targeted expression of wild-type FGFR2 appended with nuclear and nucleolar localization signals. CONCLUSIONS: Our results indicate that the bent long bones in BBDS arise from disruptions in musculoskeletal integration and that increased nuclear and nucleolar localization of FGFR2 plays a mechanistic role in the disease phenotype. 248:233-246, 2019. © 2018 Wiley Periodicals, Inc.


Assuntos
Núcleo Celular/química , Extremidades/crescimento & desenvolvimento , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Animais , Doenças do Desenvolvimento Ósseo/genética , Embrião de Galinha , Deformidades Congênitas dos Membros/genética , Fenótipo , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia
15.
Calcif Tissue Int ; 104(2): 201-206, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30341591

RESUMO

Nephronectin (Npnt), an extracellular matrix protein, is known to be a ligand of integrin α8ß1, and it has also been known to play critical roles as various organs. In the present study, elevated extracellular inorganic phosphate (Pi) strongly inhibited the expression of Npnt in MC3T3-E1 cells, while the existence of extracellular calcium (Ca) was indispensable for its effect. Furthermore, Pi-induced inhibition of Npnt gene expression was recovered by inhibitors of both sodium-dependent Pi transporter (Pit) and fibroblast growth factor receptors (Fgfrs). These results demonstrated that Npnt gene expression is regulated by extracellular Pi via Pit and Fgfrs.


Assuntos
Proteínas da Matriz Extracelular/genética , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Fosfatos/farmacologia , Células 3T3 , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Proteínas da Matriz Extracelular/metabolismo , Camundongos , Proteínas de Transporte de Fosfato/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
16.
Yi Chuan ; 40(7): 515-524, 2018 Jul 20.
Artigo em Chinês | MEDLINE | ID: mdl-30021715

RESUMO

The inner ear is a complex sensory organ that detects sound and mediates balance. During inner ear development, fibroblast growth factor (FGF) signaling pathway is involved in the induction of otic placode, cell fate determination of statoacoustic ganglion (SAG) neurons, and epithelial differentiation of the Corti organ. FGF signaling initiates the regulatory network of otic genes in the early development of inner ear, and induces the formation of pre-placodal region and the otic placode. The specification of the neuroblast ventral to the otic vesicle could be promoted by the normally-expressed FGF, and inhibited by excessive FGF5 secreted by mature SAG neurons, which could form a negative feedback loop and stabilize the SAG cell identity. The expression of FGF20 is regulated by the Notch signaling pathway and implicated in the differentiation of hair cells and supporting cells in the prosensory epithelium. FGF8 secreted by hair cells could regulate the differentiation of partial supporting cells into pillar cells. Abnormal FGF signaling in humans could lead to different kinds of deafness-related genetic diseases. In addition, it is noteworthy that FGF signaling pathway plays an important role in hair cell regeneration and induction from stem cells in lower vertebrates. In this review, we summarize recent advancements on roles of the FGF signaling pathway in inner ear development and hair cell regeneration, and lay a theoretical foundation for elucidating the regulatory mechanisms of FGF signal pathway in hair cell regeneration.


Assuntos
Orelha Interna/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais , Animais , Humanos , Regeneração
17.
J Neurosci ; 37(50): 12094-12105, 2017 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-29097598

RESUMO

Fibroblast growth factors (FGFs) and FGF receptors (FGFRs) are known for their potent effects on cell proliferation/differentiation and cortical patterning in the developing brain. However, little is known regarding the roles of FGFs/FGFRs in cortical circuit formation. Here we show that Fgfr1/2/3 and Fgf7/9/10/22 mRNAs are expressed in the developing primary somatosensory (S1) barrel cortex. Barrel cortex layer IV spiny stellate cells (bSCs) are the primary recipients of ascending sensory information via thalamocortical axons (TCAs). Detail quantification revealed distinctive phases for bSC dendritogenesis: orienting dendrites toward TCAs, adding de novo dendritic segments, and elongating dendritic length, while maintaining dendritic patterns. Deleting Fgfr1/2/3 in bSCs had minimal impact on dendritic polarity but transiently increased the number of dendritic segments. However, 6 d later, FGFR1/2/3 loss of function reduced dendritic branch numbers. These data suggest that FGFs/FGFRs have a role in stabilizing dendritic patterning. Depolarization of cultured mouse cortical neurons upregulated the levels of several Fgf/Fgfr mRNAs within 2 h. In vivo, within 6 h of systemic kainic acid administration at postnatal day 6, mRNA levels of Fgf9, Fgf10, Fgfr2c, and Fgfr3b in S1 cortices were enhanced, and this was accompanied by exuberant dendritogenesis of bSCs by 24 h. Deleting Fgfr1/2/3 abolished kainic acid-induced bSC dendritic overgrowth. Finally, FGF9/10 gain of function also resulted in extensive dendritogenesis. Together, our data suggest that FGFs/FGFRs can be regulated by glutamate transmission to modulate/stabilize bSC dendritic complexity. Both male and female mice were used for our study.SIGNIFICANCE STATEMENT Glutamatergic transmission plays critical roles in cortical circuit formation. Its dysregulation has been proposed as a core factor in the etiology of many neurological diseases. We found that excessive glutamate transmission upregulated mRNA expression of Fgfrs and their ligands Fgfs Deleting Fgfr1/2/3 not only impaired bSC dendritogenesis but also abolished glutamate transmission-induced dendritic overgrowth. Overexpressing FGF9 or FGF10 in cortical glutamatergic neurons results in excessive dendritic outgrowth within 24 h, resembling the changes induced by excessive glutamate transmission. Our findings provide strong evidence for the physiological role of fibroblast growth factors (FGFs) and FGF receptors (FGFRs) in establishing and maintaining cortical circuits. Perturbing the expression levels of FGFs/FGFRs by excessive glutamatergic neurotransmission could lead to abnormal neuronal circuits, which may contribute to neurological and psychiatric disease.


Assuntos
Dendritos/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurogênese , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Córtex Somatossensorial/embriologia , Vibrissas/inervação , Animais , Células Cultivadas , Convulsivantes/toxicidade , Eletroporação , Feminino , Fatores de Crescimento de Fibroblastos/genética , Mutação com Ganho de Função , Ácido Caínico/toxicidade , Mutação com Perda de Função , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos Transgênicos , Neurogênese/efeitos dos fármacos , Neurônios/citologia , Gravidez , Receptores de Fatores de Crescimento de Fibroblastos/deficiência , Receptores de Fatores de Crescimento de Fibroblastos/genética , Proteínas Recombinantes de Fusão/metabolismo , Córtex Somatossensorial/citologia , Córtex Somatossensorial/efeitos dos fármacos
18.
Cancer Cell ; 32(4): 474-489.e6, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-29017058

RESUMO

Androgen receptor (AR) signaling is a distinctive feature of prostate carcinoma (PC) and represents the major therapeutic target for treating metastatic prostate cancer (mPC). Though highly effective, AR antagonism can produce tumors that bypass a functional requirement for AR, often through neuroendocrine (NE) transdifferentiation. Through the molecular assessment of mPCs over two decades, we find a phenotypic shift has occurred in mPC with the emergence of an AR-null NE-null phenotype. These "double-negative" PCs are notable for elevated FGF and MAPK pathway activity, which can bypass AR dependence. Pharmacological inhibitors of MAPK or FGFR repressed the growth of double-negative PCs in vitro and in vivo. Our results indicate that FGF/MAPK blockade may be particularly efficacious against mPCs with an AR-null phenotype.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Neoplasias da Próstata/patologia , Receptores Androgênicos/fisiologia , Transdução de Sinais/fisiologia , Antagonistas de Androgênios/uso terapêutico , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Humanos , Proteína 1 Inibidora de Diferenciação/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Metástase Neoplásica , Neoplasias da Próstata/tratamento farmacológico , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia
19.
Crit Rev Oncol Hematol ; 113: 256-267, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28427515

RESUMO

INTRODUCTION: The Fibroblast Growth Factor Receptor (FGFR) family consists of Tyrosine Kinase Receptors (TKR) involved in several biological functions. Recently, alterations of FGFR have been reported to be important for progression and development of several cancers. In this setting, different studies are trying to evaluate the efficacy of different therapies targeting FGFR. AREAS COVERED: This review summarizes the current status of treatments targeting FGFR, focusing on the trials that are evaluating the FGFR profile as inclusion criteria: Multi-Target, Pan-FGFR Inhibitors and anti-FGF (Fibroblast Growth Factor)/FGFR Monoclonal Antibodies. EXPERT OPINION: Most of the TKR share intracellular signaling pathways; therefore, cancer cells tend to overcome the inhibition of one tyrosine kinase receptor by activating another. The future of TKI (Tyrosine Kinase Inhibitor) therapy will potentially come from multi-targeted TKIs that target different TKR simultaneously. It is crucial to understand the interaction of the FGF-FGFR axis with other known driver TKRs. Based on this, it is possible to develop therapeutic strategies targeting multiple connected TKRs at once. One correct step in this direction is the reassessment of multi target inhibitors considering the FGFR status of the tumor. Another opportunity arises from assessing the use of FGFR TKI on patients harboring FGFR alterations.


Assuntos
Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/fisiologia , Fusão Gênica , Humanos , Terapia de Alvo Molecular , Mutação , Neoplasias/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/fisiologia
20.
Dev Dyn ; 246(4): 344-352, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28198118

RESUMO

Fibroblast growth factors (FGFs) are a family of conserved peptides that play an important role in the development, homeostasis, and repair processes of many organ systems, including the gastrointestinal tract. All four FGF receptors and several FGF ligands are present in the intestine. They play important roles in controlling cell proliferation, differentiation, epithelial cell restitution, and stem cell maintenance. Several FGFs have also been proven to be protective against gastrointestinal diseases such as inflammatory bowel diseases or to aid in regeneration after intestinal loss associated with short bowel syndrome. Herein, we review the multifaceted actions of canonical FGFs in intestinal development, homeostasis, and repair in rodents and humans. Developmental Dynamics 246:344-352, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Trato Gastrointestinal/fisiologia , Regeneração , Animais , Gastroenteropatias/metabolismo , Gastroenteropatias/prevenção & controle , Trato Gastrointestinal/química , Trato Gastrointestinal/metabolismo , Humanos , Intestinos/citologia , Intestinos/crescimento & desenvolvimento , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Roedores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...