Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.316
Filtrer
1.
Front Immunol ; 15: 1390453, 2024.
Article de Anglais | MEDLINE | ID: mdl-38962005

RÉSUMÉ

Fibroblast growth factors (FGFs) are a versatile family of peptide growth factors that are involved in various biological functions, including cell growth and differentiation, embryonic development, angiogenesis, and metabolism. Abnormal FGF/FGF receptor (FGFR) signaling has been implicated in the pathogenesis of multiple diseases such as cancer, metabolic diseases, and inflammatory diseases. It is worth noting that macrophage polarization, which involves distinct functional phenotypes, plays a crucial role in tissue repair, homeostasis maintenance, and immune responses. Recent evidence suggests that FGF/FGFR signaling closely participates in the polarization of macrophages, indicating that they could be potential targets for therapeutic manipulation of diseases associated with dysfunctional macrophages. In this article, we provide an overview of the structure, function, and downstream regulatory pathways of FGFs, as well as crosstalk between FGF signaling and macrophage polarization. Additionally, we summarize the potential application of harnessing FGF signaling to modulate macrophage polarization.


Sujet(s)
Facteurs de croissance fibroblastique , Macrophages , Récepteur facteur croissance fibroblaste , Transduction du signal , Humains , Macrophages/immunologie , Macrophages/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Animaux , Récepteur facteur croissance fibroblaste/métabolisme , Activation des macrophages/immunologie , Inflammation/immunologie , Inflammation/métabolisme
2.
Cancer Lett ; 595: 216997, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-38801887

RÉSUMÉ

Anti-FGFR treatment for cholangiocarcinoma (CCA) with fibroblast growth factor receptor (FGFR) alteration is a promising treatment option. Since the antitumor mechanisms of anti-FGFR inhibitors and conventional cytotoxic drugs differ, synergistic effects can be possible. This study aimed to evaluate the efficacy of the combined administration of gemcitabine (GEM) and pemigatinib in CCA cells with FGFR2 alterations. To simulate the treatment for patients with 3 kinds of CCA, chemonaïve CCA with activation of the FGF pathway, chemo-resistant CCA with activation of the FGF pathway, and CCA without FGF pathway activation (as controls), we evaluated 3 different CCA cell lines, CCLP-1 (with a FGFR2 fusion mutation), CCLP-GR (GEM-resistant cells established from CCLP-1), and HuCCT1 (without FGFR mutations). There was no significant difference between CCLP-1 and HuCCT1 in GEM suspensibility (IC50 = 19.3, 22.6 mg/dl, p = 0.1187), and the drug sensitivity to pemigatinib did not differ between CCLP-1 and CCLP-GR (IC50 = 7.18,7.60 nM, p = 0.3089). Interestingly, only CCLP-1 showed a synergistic effect with combination therapy consisting of GEM plus pemigatinib in vitro and in vivo. In a comparison of the reaction to GEM exposure, only CCLP-1 cells showed an increase in the activation of downstream proteins in the FGF pathway, especially FRS2 and ERK. In association with this reaction, cell cycle and mitosis were increased with GEM exposure in CCLP-1, but HuCCT1/CCLP-GR did not show this reaction. Our results suggested that combination therapy with GEM plus pemigatinib is a promising treatment for chemonaïve patients with CCA with activation of the FGF pathway.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs des canaux biliaires , Cholangiocarcinome , Désoxycytidine , Synergie des médicaments , , Pyrimidines , Récepteur FGFR2 , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Cholangiocarcinome/traitement médicamenteux , Cholangiocarcinome/anatomopathologie , Cholangiocarcinome/génétique , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Désoxycytidine/administration et posologie , Animaux , Tumeurs des canaux biliaires/traitement médicamenteux , Tumeurs des canaux biliaires/anatomopathologie , Tumeurs des canaux biliaires/génétique , Lignée cellulaire tumorale , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Pyrimidines/pharmacologie , Pyrimidines/administration et posologie , Récepteur FGFR2/antagonistes et inhibiteurs , Récepteur FGFR2/métabolisme , Récepteur FGFR2/génétique , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs de croissance fibroblastique/métabolisme , Facteurs de croissance fibroblastique/génétique , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/métabolisme , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Mutation , Apoptose/effets des médicaments et des substances chimiques , Morpholines , Pyrroles
3.
Cytokine Growth Factor Rev ; 77: 39-55, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38719671

RÉSUMÉ

Fibroblast growth factors (FGFs) and their receptors (FGFRs) constitute plasma-membrane localized signaling hubs that transmit signals from the extracellular environment to the cell interior, governing pivotal cellular processes like motility, metabolism, differentiation, division and death. FGF/FGFR signaling is critical for human body development and homeostasis; dysregulation of FGF/FGFR units is observed in numerous developmental diseases and in about 10% of human cancers. Glycosylation is a highly abundant posttranslational modification that is critical for physiological and pathological functions of the cell. Glycosylation is also very common within FGF/FGFR signaling hubs. Vast majority of FGFs (15 out of 22 members) are N-glycosylated and few FGFs are O-glycosylated. Glycosylation is even more abundant within FGFRs; all FGFRs are heavily N-glycosylated in numerous positions within their extracellular domains. A growing number of studies points on the multiple roles of glycosylation in fine-tuning FGF/FGFR signaling. Glycosylation modifies secretion of FGFs, determines their stability and affects interaction with FGFRs and co-receptors. Glycosylation of FGFRs determines their intracellular sorting, constitutes autoinhibitory mechanism within FGFRs and adjusts FGF and co-receptor recognition. Sugar chains attached to FGFs and FGFRs constitute also a form of code that is differentially decrypted by extracellular lectins, galectins, which transform FGF/FGFR signaling at multiple levels. This review focuses on the identified functions of glycosylation within FGFs and FGFRs and discusses their relevance for the cell physiology in health and disease.


Sujet(s)
Facteurs de croissance fibroblastique , Récepteur facteur croissance fibroblaste , Transduction du signal , Humains , Glycosylation , Facteurs de croissance fibroblastique/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme , Animaux , Maturation post-traductionnelle des protéines
4.
Curr Opin Oncol ; 36(4): 320-325, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38726837

RÉSUMÉ

PURPOSE OF REVIEW: This review is timely and relevant due to the increasing recognition of the significance of the fibroblast growth factor receptor (FGFR) family in cancer biology. Understanding the role of FGFRs and their dysregulation in various cancers is crucial for developing targeted therapies and improving patient outcomes. RECENT FINDINGS: The review highlights the importance of the FGFR family in cellular processes such as growth, proliferation, and survival. It discusses how abnormalities in FGFR2, including overexpression, gene amplification, and other genetic alterations, contribute to cancer progression, particularly in gastro-intestinal cancers. The paper also emphasizes the promising results of FGFR-targeted therapies, especially tyrosine kinase inhibitors, in certain cancers such as cholangiocarcinoma and oesophagogastric cancers. SUMMARY: The findings underscore the potential of FGFR-targeted therapies in treating cancers with FGFR dysregulation. However, the review also addresses the challenges associated with these therapies, including toxicities and mechanisms of resistance. Understanding these complexities is essential for optimizing the efficacy of FGFR-targeted treatments and improving patient outcomes in clinical practice and research efforts.


Sujet(s)
Tumeurs gastro-intestinales , Récepteur facteur croissance fibroblaste , Humains , Tumeurs gastro-intestinales/traitement médicamenteux , Tumeurs gastro-intestinales/génétique , Tumeurs gastro-intestinales/métabolisme , Tumeurs gastro-intestinales/anatomopathologie , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/métabolisme , Thérapie moléculaire ciblée , Inhibiteurs de protéines kinases/usage thérapeutique , Récepteur FGFR2/antagonistes et inhibiteurs , Récepteur FGFR2/génétique , Récepteur FGFR2/métabolisme , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie
5.
Pharmacol Ther ; 259: 108669, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38795981

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal disease for which therapeutic options are limited, with an unmet need to identify new therapeutic targets. IPF is thought to be the consequence of repeated microlesions of the alveolar epithelium, leading to aberrant epithelial-mesenchymal communication and the accumulation of extracellular matrix proteins. The reactivation of developmental pathways, such as Fibroblast Growth Factors (FGFs), is a well-described mechanism during lung fibrogenesis. Secreted FGFs with local paracrine effects can either exert an anti-fibrotic or a pro-fibrotic action during this pathological process through their FGF receptors (FGFRs) and heparan sulfate residues as co-receptors. Among FGFs, endocrine FGFs (FGF29, FGF21, and FGF23) play a central role in the control of metabolism and tissue homeostasis. They are characterized by a low affinity for heparan sulfate, present in the cell vicinity, allowing them to have endocrine activity. Nevertheless, their interaction with FGFRs requires the presence of mandatory co-receptors, alpha and beta Klotho proteins (KLA and KLB). Endocrine FGFs are of growing interest for their anti-fibrotic action during liver, kidney, or myocardial fibrosis. Innovative therapies based on FGF19 or FGF21 analogs are currently being studied in humans during liver fibrosis. Recent data report a similar anti-fibrotic action of endocrine FGFs in the lung, suggesting a systemic regulation of the pulmonary fibrotic process. In this review, we summarize the current knowledge on the protective effect of endocrine FGFs during the fibrotic processes, with a focus on pulmonary fibrosis.


Sujet(s)
Facteurs de croissance fibroblastique , Humains , Facteurs de croissance fibroblastique/métabolisme , Animaux , Facteur-23 de croissance des fibroblastes , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Récepteur facteur croissance fibroblaste/métabolisme
6.
Bull Exp Biol Med ; 176(5): 576-580, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38724808

RÉSUMÉ

We performed a comparative in vitro study of the involvement of NF-κB, PI3K, cAMP, ERK1/2, p38, JAKs, STAT3, JNK, and p53-dependent intracellular signaling in the functioning of neural stem cells (NSC) under the influence of basic fibroblast growth factor (FGF) and FGF receptor agonist, diterpene alkaloid songorine. The significant differences in FGFR-mediated intracellular signaling in NSC were revealed for these ligands. In both cases, stimulation of progenitor cell proliferation occurs with the participation of NF-κB, PI3K, ERK1/2, JAKs, and STAT3, while JNK and p53, on the contrary, inhibit cell cycle progression. However, under the influence of songorin, cAMP- and p38-mediated cascades are additionally involved in the transmission of the NSC division-activating signal. In addition, unlike FGF, the alkaloid stimulates progenitor cell differentiation by activating ERK1/2, p38, JNK, p53, and STAT3.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Diterpènes , Cellules souches neurales , Récepteur facteur croissance fibroblaste , Facteur de transcription STAT-3 , Transduction du signal , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Animaux , Facteur de transcription STAT-3/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme , Récepteur facteur croissance fibroblaste/agonistes , Transduction du signal/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Diterpènes/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Facteur de croissance fibroblastique de type 2/métabolisme , Facteur de croissance fibroblastique de type 2/pharmacologie , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/agonistes , Phosphatidylinositol 3-kinases/métabolisme , Alcaloïdes/pharmacologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Janus kinases/métabolisme , AMP cyclique/métabolisme , Cellules cultivées , Rats
7.
Front Biosci (Landmark Ed) ; 29(5): 184, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38812320

RÉSUMÉ

This review article explores the intricate correlation between growth factors and bone metastases, which play a crucial role in the development of several types of malignancies, namely breast, prostate, lung, and renal cancers. The focal point of our discussion is on crucial receptors for growth factors, including Epidermal Growth Factor Receptor (EGFR), Transforming Growth Factor-ß (TGFß), Vascular Endothelial Growth Factor Receptor (VEGFR), and Fibroblast Growth Factor Receptor (FGFR). These receptors, which are essential for cellular activities including growth, differentiation, and survival, have important involvement in the spread of cancer and the interactions between tumors and the bone environment. We discuss the underlying mechanisms of bone metastases, with a specific emphasis on the interaction between growth factor receptors and the bone microenvironment. EGFR signaling specifically enhances the process of osteoclast development and the formation of osteolytic lesions, especially in breast and lung malignancies. TGFß receptors have a role in both osteolytic and osteoblastic metastases by releasing TGFß, which attracts cancer cells and promotes bone remodeling. This is a crucial element in the spread of prostate cancer to the bones. The functions of FGFR and VEGFR in the processes of bone formation and tumor angiogenesis, respectively, highlight the complex and diverse nature of these interactions. The review emphasizes the possibility of targeted therapeutics targeting these receptors to interrupt the cycle of tumor development and bone degradation. Therapeutic approaches include focusing on the VEGF/VEGFR, EGF/EGFR, FGF/FGFR, and TGFß/TGFßR pathways. These include a variety of compounds, such as small molecule inhibitors and monoclonal antibodies, which have shown potential to interfere with tumor-induced alterations in bone. The text discusses clinical trials and preclinical models, offering insights into the effectiveness and constraints of various treatments. Ultimately, this study provides a succinct but thorough summary of the present knowledge and treatment strategies focused on growth factor receptors in bone metastases. This highlights the significance of comprehending the signaling of growth factor receptors in the microenvironment where tumors spread to the bones, as well as the possibility of using targeted therapies to enhance the results for cancer patients with bone metastases. The advancement of treating bone metastases hinges on the development of treatments that specifically target the intricate relationships between malignancies and bone.


Sujet(s)
Tumeurs osseuses , Humains , Tumeurs osseuses/secondaire , Tumeurs osseuses/métabolisme , Récepteur facteur croissance/métabolisme , Transduction du signal , Facteur de croissance transformant bêta/métabolisme , Récepteurs ErbB/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/métabolisme , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Animaux , Récepteurs aux facteurs de croissance endothéliale vasculaire/métabolisme , Récepteurs aux facteurs de croissance endothéliale vasculaire/antagonistes et inhibiteurs
8.
Front Immunol ; 15: 1327416, 2024.
Article de Anglais | MEDLINE | ID: mdl-38638441

RÉSUMÉ

In previous studies, we showed that fibroblast growth factor receptors (FGFRs) contribute to inflammatory mediator output from primary rhesus microglia in response to live Borrelia burgdorferi. We also demonstrated that non-viable B. burgdorferi can be as pathogenic as live bacteria, if not more so, in both CNS and PNS tissues. In this study we assessed the effect of live and non-viable B. burgdorferi in inducing FGFR expression from rhesus frontal cortex (FC) and dorsal root ganglion (DRG) tissue explants as well as their neuronal/astrocyte localization. Specific FGFR inhibitors were also tested for their ability to attenuate inflammatory output and apoptosis in response to either live or non-viable organisms. Results show that in the FC, FGFR2 was the most abundantly expressed receptor followed by FGFR3 and FGFR1. Non-viable B. burgdorferi significantly upregulated FGFR3 more often than live bacteria, while the latter had a similar effect on FGFR1, although both treatments did affect the expressions of both receptors. FGFR2 was the least modulated in the FC tissues by the two treatments. FGFR1 expression was more prevalent in astrocytes while FGFR2 and FGFR3 showed higher expression in neurons. In the DRG, all three receptor expressions were also seen, but could not be distinguished from medium controls by immunofluorescence. Inhibition of FGFR1 by PD166866 downregulated both inflammation and apoptosis in both FC and DRG in response to either treatment in all the tissues tested. Inhibition of FGFR1-3 by AZD4547 similarly downregulated both inflammation and apoptosis in both FC and DRG in response to live bacteria, while with sonicated remnants, this effect was seen in one of the two FC tissues and 2 of 3 DRG tissues tested. CCL2 and IL-6 were the most downregulated mediators in the FC, while in the DRG it was CXCL8 and IL-6 in response to FGFR inhibition. Downregulation of at least two of these three mediators was observed to downregulate apoptosis levels in general. We show here that FGFR inhibition can be an effective anti-inflammatory treatment in antibiotic refractive neurological Lyme. Alternatively, two biologics may be needed to effectively curb neuroinflammation and pathology in the CNS and PNS.


Sujet(s)
Borrelia burgdorferi , Humains , Interleukine-6/métabolisme , Inflammation/métabolisme , Neurones/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme
9.
Dalton Trans ; 53(18): 7682-7693, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38573236

RÉSUMÉ

Dysregulation of Fibroblast Growth Factor Receptors (FGFRs) signaling has been associated with breast cancer, yet employing FGFR-targeted delivery systems to improve the efficacy of cytotoxic agents is still sparsely exploited. Herein, we report four new bi-functional ruthenium-peptide conjugates (RuPCs) with FGFR-targeting and pH-dependent releasing abilities, envisioning the selective delivery of cytotoxic Ru complexes to FGFR(+)-breast cancer cells, and controlled activation at the acidic tumoral microenvironment. The antiproliferative potential of the RuPCs and free Ru complexes was evaluated in four breast cancer cell lines with different FGFR expression levels (SKBR-3, MDA-MB-134-VI, MCF-7, and MDA-MB-231) and in human dermal fibroblasts (HDF), at pH 6.8 and pH 7.4 aimed at mimicking the tumor microenvironment and normal tissues/bloodstream pHs, respectively. The RuPCs showed higher cytotoxicity in cells with higher level of FGFR expression at acidic pH. Additionally, RuPCs showed up to 6-fold higher activity in the FGFR(+) breast cancer lines compared to the normal cell line. The release profile of Ru complexes from RuPCs corroborates the antiproliferative effects observed. Remarkably, the cytotoxicity and releasing ability of RuPCs were shown to be strongly dependent on the conjugation of the peptide position in the Ru complex. Complementary molecular dynamic simulations and computational calculations were performed to help interpret these findings at the molecular level. In summary, we identified a lead bi-functional RuPC that holds strong potential as a FGFR-targeted chemotherapeutic agent.


Sujet(s)
Antinéoplasiques , Tumeurs du sein , Peptides , Récepteur facteur croissance fibroblaste , Ruthénium , Femelle , Humains , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Complexes de coordination/composition chimique , Complexes de coordination/pharmacologie , Complexes de coordination/synthèse chimique , Tests de criblage d'agents antitumoraux , Concentration en ions d'hydrogène , Peptides/composition chimique , Peptides/pharmacologie , Peptides/usage thérapeutique , Récepteur facteur croissance fibroblaste/métabolisme , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Ruthénium/composition chimique , Ruthénium/pharmacologie , Ruthénium/usage thérapeutique
10.
Int J Biol Macromol ; 267(Pt 1): 131524, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38608977

RÉSUMÉ

As our grasp of cancer genomics deepens, we are steadily progressing towards the domain of precision medicine, where targeted therapy stands out as a revolutionary breakthrough in the landscape of cancer therapeutics. The fibroblast growth factor receptors (FGFR) pathway has been unveiled as a fundamental instigator in the pathophysiological mechanisms underlying breast carcinoma, paving the way for the exhilarating development of precision-targeted therapeutics. In the pursuit of exploring inhibitors that specifically target the FGFR signaling pathways, a multitude of kinase inhibitors targeting FGFR has been assiduously engineered to address the heterogeneous landscape of human malignancies. This review offers an exhaustive exploration of aberrations within the FGFR pathway and their functional implications in breast cancer. Additionally, we delve into cutting-edge therapeutic approaches for the treatment of breast cancer patients bearing FGFR alterations and the management of toxicity associated with FGFR inhibitors. Furthermore, our contemplation of the evolution of cutting-edge FGFR inhibitors foresees their potential to spearhead innovative therapeutic approaches in the ongoing combat against cancer.


Sujet(s)
Tumeurs du sein , Inhibiteurs de protéines kinases , Récepteur facteur croissance fibroblaste , Transduction du signal , Humains , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Femelle , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Thérapie moléculaire ciblée , Animaux
11.
Front Endocrinol (Lausanne) ; 15: 1379231, 2024.
Article de Anglais | MEDLINE | ID: mdl-38638139

RÉSUMÉ

Receptor tyrosine kinases (RTKs) mediate the actions of growth factors in metazoans. In decapod crustaceans, RTKs are implicated in various physiological processes, such molting and growth, limb regeneration, reproduction and sexual differentiation, and innate immunity. RTKs are organized into two main types: insulin receptors (InsRs) and growth factor receptors, which include epidermal growth factor receptor (EGFR), fibroblast growth factor receptor (FGFR), vascular endothelial growth factor receptor (VEGFR), and platelet-derived growth factor receptor (PDGFR). The identities of crustacean RTK genes are incomplete. A phylogenetic analysis of the CrusTome transcriptome database, which included all major crustacean taxa, showed that RTK sequences segregated into receptor clades representing InsR (72 sequences), EGFR (228 sequences), FGFR (129 sequences), and PDGFR/VEGFR (PVR; 235 sequences). These four receptor families were distinguished by the domain organization of the extracellular N-terminal region and motif sequences in the protein kinase catalytic domain in the C-terminus or the ligand-binding domain in the N-terminus. EGFR1 formed a single monophyletic group, while the other RTK sequences were divided into subclades, designated InsR1-3, FGFR1-3, and PVR1-2. In decapods, isoforms within the RTK subclades were common. InsRs were characterized by leucine-rich repeat, furin-like cysteine-rich, and fibronectin type 3 domains in the N-terminus. EGFRs had leucine-rich repeat, furin-like cysteine-rich, and growth factor IV domains. N-terminal regions of FGFR1 had one to three immunoglobulin-like domains, whereas FGFR2 had a cadherin tandem repeat domain. PVRs had between two and five immunoglobulin-like domains. A classification nomenclature of the four RTK classes, based on phylogenetic analysis and multiple sequence alignments, is proposed.


Sujet(s)
Furine , Insuline , Furine/génétique , Phylogenèse , Insuline/génétique , Transcriptome , Cystéine , Leucine/génétique , Facteur de croissance endothéliale vasculaire de type A/génétique , Récepteurs à activité tyrosine kinase/génétique , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs ErbB/métabolisme , Récepteur facteur croissance fibroblaste/génétique , Récepteur facteur croissance fibroblaste/métabolisme , Analyse de profil d'expression de gènes , Tyrosine
12.
Eur J Pharmacol ; 971: 176521, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38522639

RÉSUMÉ

Maintaining blood-brain barrier (BBB) integrity is critical components of therapeutic approach for ischemic stroke. Fibroblast growth factor 17 (FGF17), a member of FGF8 superfamily, exhibits the strongest expression throughout the wall of all major arteries during development. However, its molecular action and potential protective role on brain endothelial cells after stroke remains unclear. Here, we observed reduced levels of FGF17 in the serum of patients with ischemic stroke, as well as in the brains of mice subjected to middle cerebral artery occlusion (MCAO) injury and oxygen-glucose deprivation/reoxygenation (OGD/R)-induced brain microvascular endothelial cells (bEnd.3) cells. Moreover, treatment with exogenous recombinant human FGF17 (rhFGF17) decreased infarct volume, improved neurological deficits, reduced Evans Blue leakage and upregulated the expression of tight junctions in MCAO-injured mice. Meanwhile, rhFGF17 increased cell viability, enhanced trans-endothelial electrical resistance, reduced sodium fluorescein leakage, and alleviated reactive oxygen species (ROS) generation in OGD/R-induced bEnd.3 cells. Mechanistically, the treatment with rhFGF17 resulted in nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear accumulation and upregulation of heme oxygenase-1 (HO-1) expression. Additionally, based on in-vivo and in-vitro research, rhFGF17 exerted protective effects against ischemia/reperfusion (I/R) -induced BBB disruption and endothelial cell apoptosis through the activation of the FGF receptor 3/PI3K/AKT signaling pathway. Overall, our findings indicated that FGF17 may hold promise as a novel therapeutic strategy for ischemic stroke patients.


Sujet(s)
Encéphalopathie ischémique , Accident vasculaire cérébral ischémique , Lésion d'ischémie-reperfusion , Rats , Humains , Souris , Animaux , Barrière hémato-encéphalique/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Cellules endothéliales , Phosphatidylinositol 3-kinases/métabolisme , Rat Sprague-Dawley , Transduction du signal , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/métabolisme , Reperfusion , Oxygène/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/prévention et contrôle , Lésion d'ischémie-reperfusion/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/complications , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Accident vasculaire cérébral ischémique/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme , Récepteur facteur croissance fibroblaste/usage thérapeutique , Facteurs de croissance fibroblastique/métabolisme
13.
Surv Ophthalmol ; 69(1): 34-41, 2024.
Article de Anglais | MEDLINE | ID: mdl-37777119

RÉSUMÉ

Fibroblast growth factor receptor (FGFR) inhibitors are an emerging class of small molecule targeted cancer drugs with promising therapeutic possibilities for a wide variety of malignancies. While ocular adverse events from FGFR inhibitors are reported in clinical trials, subsequent case studies continue to reveal new toxicities. Disease pathology affecting multiple parts of the eye has been reported, but the ocular surface and the retina are the most commonly encountered areas affected by FGFR inhibitors, manifesting as dry eye and FGFR inhibitor-associated retinopathy, respectively. Corneal thinning and melt is a rare but serious and potentially vision-threatening complication of FGFR inhibitor toxicity. Similarities between toxicities observed from other targeted cancer therapy drugs and FGFR inhibitors may help us understand underlying pathophysiological changes. The management of these adverse events requires close ophthalmologic follow-up and may require discontinuation of the offending agents in some cases.


Sujet(s)
Antinéoplasiques , Maladies de l'oeil , Humains , Récepteur facteur croissance fibroblaste/métabolisme , Antinéoplasiques/effets indésirables , Inhibiteurs de protéines kinases/effets indésirables , Maladies de l'oeil/induit chimiquement , Maladies de l'oeil/traitement médicamenteux
14.
Am J Physiol Endocrinol Metab ; 326(2): E134-E147, 2024 02 01.
Article de Anglais | MEDLINE | ID: mdl-38117265

RÉSUMÉ

Vertical sleeve gastrectomy (VSG) restores glucose homeostasis in obese mice and humans. In addition, the increased fibroblast growth factor (FGF)15/19 circulating level postsurgery has been implicated in this effect. However, the impact of FGF15/19 on pancreatic islets remains unclear. Using a diet-induced obese mice model, we demonstrate that VSG attenuates insulin hypersecretion in isolated pancreatic islets, likely due to morphological alterations in the endocrine pancreas such as reduction in islet, ß-cell, and α-cell mass. In addition, VSG relieves gene expression of endoplasmic reticulum (ER) stress and inflammation markers in islets from obese mice. Incubation of INS-1E ß-cells with serum from obese mice induced dysfunction and cell death, whereas these conditions were not induced with serum from obese mice submitted to VSG, implicating the involvement of a humoral factor. Indeed, VSG increased FGF15 circulating levels in obese mice, as well as the expression of FGF receptor 1 (Fgfr1) and its coreceptor ß-klotho (Klb), both in pancreatic islets from VSG mice and in INS-1E cells treated with the serum from these mice. Moreover, exposing INS-1E cells to an FGFR inhibitor abolished the effects of VSG serum on insulin secretion and cell death. Also, recombinant FGF19 prevents INS-1E cells from dysfunction and death induced by serum from obese mice. These findings indicate that the amelioration of glucose-insulin homeostasis promoted by VSG is mediated, at least in part, by FGF15/19. Therefore, approaches promoting FGF15/19 release or action may restore pancreatic islet function in obesity.NEW & NOTEWORTHY Vertical sleeve gastrectomy (VSG) decreases insulin secretion, endoplasmic reticulum (ER) stress, and inflammation in pancreatic islets from obese mice. In addition, VSG increased fibroblast growth factor (FGF)15 circulating levels in obese mice, as well as the expression of FGF receptor 1 (Fgfr1) and its coreceptor ß-klotho (Klb), both in pancreatic islets from VSG mice and in INS-1E ß-cells treated with the serum from these mice. Serum from operated mice protects INS-1E cells from dysfunction and apoptosis, which was mediated by FGF15/19.


Sujet(s)
Cellules à insuline , Insuline , Souris , Humains , Animaux , Insuline/métabolisme , Souris obèse , Cellules à insuline/métabolisme , Glucose/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Gastrectomie , Inflammation/métabolisme , Homéostasie
15.
Chin J Physiol ; 66(5): 295-305, 2023.
Article de Anglais | MEDLINE | ID: mdl-37929340

RÉSUMÉ

The morbidity and mortality of prostate cancer are increasing year by year, and the survival rate of prostate cancer patients after treatment is low. Therefore, investigating the molecular mechanism underlying prostate cancer is crucial for developing effective treatments. Recent studies have shown the important role of long-chain non-coding RNAs (lncRNAs) in tumorigenesis. VPS9D1-AS1 can modulate the progression of multiple cancers, but its molecular action mechanism in prostate cancer remains unknown. This study, therefore, intended to investigate the regulatory mechanism of VPS9D1-AS1 in prostate cancer. First, differentially expressed lncRNAs in prostate cancer were identified through bioinformatics approaches. The target lncRNA for the study was determined by reviewing the relevant literature and its downstream miRNA/mRNA axis was uncovered. Then, quantitative reverse transcription polymerase chain reaction was introduced to assess the expression of VPS9D1-AS1, miR-187-3p, and fibroblast growth factor receptor-like 1 (FGFRL1) at a cellular level, and Western blot was conducted to assess the protein level of FGFRL1 in cells. The results indicated that VPS9D1-AS1 and FGFRL1 were highly expressed in prostate cancer while miR-187-3p was less expressed. Besides, MTT, colony formation, wound healing, and cell invasion assays showed that silencing VPS9D1-AS1 inhibited the viability, migration ability, and invasion ability of prostate cancer cells. Dual-luciferase assay and RNA binding protein immunoprecipitation assay were performed to explore the interplay of miR-187-3p and VPS9D1-AS1 or FGFRL1. The results showed that VPS9D1-AS1 could sponge miR-187-3p, and FGFRL1 could serve as a direct target of miR-187-3p. Moreover, combined with the results of the rescue experiment, VPS9D1-AS1 was found to upregulate FGFRL1 by competitively sponging miR-187-3p to accelerate the malignant behaviors of prostate cancer cells. In conclusion, VPS9D1-AS1 could promote the phenotype progression of prostate cancer cells through targeting the miR-187-3p/FGFRL1 axis, and it has the potential to be a target for prostate cancer patients.


Sujet(s)
microARN , Tumeurs de la prostate , ARN long non codant , Mâle , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Récepteur facteur croissance fibroblaste/génétique , Récepteur facteur croissance fibroblaste/métabolisme , Prolifération cellulaire , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , microARN/génétique , microARN/métabolisme , Tumeurs de la prostate/génétique , Régulation de l'expression des gènes tumoraux
16.
Sci Rep ; 13(1): 20779, 2023 11 27.
Article de Anglais | MEDLINE | ID: mdl-38012225

RÉSUMÉ

Emmetropization, a natural process of ocular elongation, is closely associated with scleral remodeling. The Fibroblast growth factor-2 (FGF-2) was reported involved in scleral remodeling in myopia models. Herein, we aimed to investigate the role of scleral fibroblast-to-myofibroblast differentiation and FGF-2 in scleral remodeling during maturation. Our findings revealed that the posterior scleral fibroblasts (SFs) from mature guinea pigs exhibit increased stiffness compared to those from young guinea pigs. Moreover, mature SFs displayed decreased cell proliferation but increased levels of α-SMA, matrix metalloproteinase 2 (MMP2), and collagen 1, when compared to young SFs. Additionally, the mRNA expression of scleral Fgf-2, Fgf receptor 1 (Fgfr1), Fgfr2, Fgfr3, and Fgfr4 was increased in mature SFs. Notably, exogenous FGF-2 showed increased cell proliferation and led to decreased expression of α-SMA, MMP2, and collagen 1 in mature SFs. Overall, our findings highlight the influence of maturation on SFs from posterior scleral shells, resulting in increased stiffness and the manifestation of fibroblast-to-myofibroblast differentiation during development. Exogenous FGF-2 increased cell proliferation and reversed the age-related fibroblast-to-myofibroblast differentiation, suggesting a potential role of FGF-2 in regulating scleral remodeling.


Sujet(s)
Facteur de croissance fibroblastique de type 2 , Matrix metalloproteinase 2 , Animaux , Cochons d'Inde , Matrix metalloproteinase 2/génétique , Différenciation cellulaire/physiologie , Prolifération cellulaire , Collagène , Récepteur facteur croissance fibroblaste/métabolisme
17.
Sci Rep ; 13(1): 20223, 2023 11 18.
Article de Anglais | MEDLINE | ID: mdl-37980453

RÉSUMÉ

Several alterations in fibroblast growth factor receptor (FGFR) genes have been found in breast cancer; however, they have not been well characterized as therapeutic targets. Futibatinib (TAS-120; Taiho) is a novel, selective, pan-FGFR inhibitor that inhibits FGFR1-4 at nanomolar concentrations. We sought to determine futibatinib's efficacy in breast cancer models. Nine breast cancer patient-derived xenografts (PDXs) with various FGFR1-4 alterations and expression levels were treated with futibatinib. Antitumor efficacy was evaluated by change in tumor volume and time to tumor doubling. Alterations indicating sensitization to futibatinib in vivo were further characterized in vitro. FGFR gene expression between patient tumors and matching PDXs was significantly correlated; however, overall PDXs had higher FGFR3-4 expression. Futibatinib inhibited tumor growth in 3 of 9 PDXs, with tumor stabilization in an FGFR2-amplified model and prolonged regression (> 110 days) in an FGFR2 Y375C mutant/amplified model. FGFR2 overexpression and, to a greater extent, FGFR2 Y375C expression in MCF10A cells enhanced cell growth and sensitivity to futibatinib. Per institutional and public databases, FGFR2 mutations and amplifications had a population frequency of 1.1%-2.6% and 1.5%-2.5%, respectively, in breast cancer patients. FGFR2 alterations in breast cancer may represent infrequent but highly promising targets for futibatinib.


Sujet(s)
Tumeurs du sein , Animaux , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Récepteur facteur croissance fibroblaste/métabolisme , Récepteur FGFR2/métabolisme , Pyrazoles , Pyrimidines/pharmacologie , Pyrroles , Récepteur FGFR1/génétique , Modèles animaux de maladie humaine
18.
J Immunother Cancer ; 11(10)2023 10.
Article de Anglais | MEDLINE | ID: mdl-37890888

RÉSUMÉ

A novel crosstalk between immunogenic and oncometabolic pathways triggered by T cell-released interferon-gamma (IFN-É£) has been recently identified. This IFN-É£-pyruvate kinase M2-ß-catenin axis relies on fibroblast growth factor 2 (FGF2) signaling in tumor cells and leads to hyperprogressive disease on immune checkpoint blockade (ICB) in preclinical models. This result underlines how IFN-É£ signaling may have distinct effects on tumor cells depending on their oncogenic and metabolic features. On the basis of these data, this study aims to explore the relationship between genomic tumor FGF2 or FGF/FGF receptor (FGFR) amplification and immunotherapy response in patients with metastatic solid cancers. We used a large genomic data set of 545 ICB-treated patients and compared outcomes between those with and without FGF2 genomic amplification. Patients with no FGF2 genomic amplification had significantly longer progression-free survival (PFS) (HR=0.55 (95% CI 0.4, 0.8); p value=0.005) and overall survival (OS) (HR=0.56 (0.3, 0.9); p value=0.02) than patients harboring an FGF2 amplification. We next questioned whether such an observation may extend to genomic amplification of the FGF/FGFR pathway. Similarly, patients with no FGF/FGFR genomic amplification had longer PFS (HR=0.71 (0.8, 0.9), p value=0.004) and OS (HR=0.77 (0.6, 1); p value=0.06). RNA sequencing analysis of tumors between the amplified and non-amplified populations showed distinct expression profiles concerning oncogenic pathways. Importantly, using a cohort of patients untreated with ICB from the The Cancer Genome Atlas, we show that FGF2 and FGF/FGFR genomic amplification were not associated with prognosis, thus demonstrating that we identified a predictive biomarker of immunotherapy resistance.


Sujet(s)
Tumeurs , Récepteur facteur croissance fibroblaste , Humains , Récepteur facteur croissance fibroblaste/génétique , Récepteur facteur croissance fibroblaste/métabolisme , Récepteur facteur croissance fibroblaste/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Facteur de croissance fibroblastique de type 2/génétique , Facteur de croissance fibroblastique de type 2/usage thérapeutique , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/anatomopathologie , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Génomique
19.
PLoS One ; 18(10): e0286040, 2023.
Article de Anglais | MEDLINE | ID: mdl-37856433

RÉSUMÉ

Activation of Map kinase/Erk signalling downstream of fibroblast growth factor (Fgf) tyrosine kinase receptors regulates gene expression required for mesoderm induction and patterning of the anteroposterior axis during Xenopus development. We have proposed that a subset of Fgf target genes are activated in the embyo in response to inhibition of a transcriptional repressor. Here we investigate the hypothesis that Cic (Capicua), which was originally identified as a transcriptional repressor negatively regulated by receptor tyrosine kinase/Erk signalling in Drosophila, is involved in regulating Fgf target gene expression in Xenopus. We characterise Xenopus Cic and show that it is widely expressed in the embryo. Fgf overexpression or ectodermal wounding, both of which potently activate Erk, reduce Cic protein levels in embryonic cells. In keeping with our hypothesis, we show that Cic knockdown and Fgf overexpression have overlapping effects on embryo development and gene expression. Transcriptomic analysis identifies a cohort of genes that are up-regulated by Fgf overexpression and Cic knockdown. We investigate two of these genes as putative targets of the proposed Fgf/Erk/Cic axis: fos and rasl11b, which encode a leucine zipper transcription factor and a ras family GTPase, respectively. We identify Cic consensus binding sites in a highly conserved region of intron 1 in the fos gene and Cic sites in the upstream regions of several other Fgf/Cic co-regulated genes, including rasl11b. We show that expression of fos and rasl11b is blocked in the early mesoderm when Fgf and Erk signalling is inhibited. In addition, we show that fos and rasl11b expression is associated with the Fgf independent activation of Erk at the site of ectodermal wounding. Our data support a role for a Fgf/Erk/Cic axis in regulating a subset of Fgf target genes during gastrulation and is suggestive that Erk signalling is involved in regulating Cic target genes at the site of ectodermal wounding.


Sujet(s)
Système de signalisation des MAP kinases , Récepteur facteur croissance fibroblaste , Animaux , Facteurs de croissance fibroblastique/génétique , Facteurs de croissance fibroblastique/métabolisme , Régulation de l'expression des gènes au cours du développement , Système de signalisation des MAP kinases/génétique , Récepteurs à activité tyrosine kinase/métabolisme , Récepteur facteur croissance fibroblaste/génétique , Récepteur facteur croissance fibroblaste/métabolisme , Transduction du signal/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Xenopus laevis/métabolisme
20.
Trends Pharmacol Sci ; 44(12): 862-864, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37778940

RÉSUMÉ

Chen et al. have derived cryogenic electron microscopy (cryo-EM) structures of signaling complexes of the endocrine hormone fibroblast growth factor 23 (FGF23) with fibroblast growth factor receptor (FGFR), α-Klotho, and heparin sulfate. These structures are asymmetric, leading to questions concerning in vivo function, and will facilitate structure-based drug design to modulate FGF23 signaling.


Sujet(s)
Facteurs de croissance fibroblastique , Protéines Klotho , Humains , Facteurs de croissance fibroblastique/composition chimique , Glucuronidase/métabolisme , Transduction du signal/physiologie , Récepteur facteur croissance fibroblaste/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...