Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(2): 102804, 2023 02.
Article in English | MEDLINE | ID: mdl-36529290

ABSTRACT

Fibroblast growth factor (FGF) is a multifunctional protein that exhibits a wide range of biological effects. Most commonly, it acts as a mitogen, but it also has regulatory, morphological, and endocrine effects. The four receptor subtypes of FGF are activated by more than 20 different FGF ligands. FGF2, one of the FGF ligands, is an essential factor for cell culture in stem cells for regenerative medicine; however, recombinant FGF2 is extremely unstable. Here, we successfully generated homobivalent agonistic single-domain antibodies (variable domain of heavy chain of heavy chain antibodies referred to as VHHs) that bind to domain III and induce activation of the FGF receptor 1 and thus transduce intracellular signaling. This agonistic VHH has similar biological activity (EC50) as the natural FGF2 ligand. Furthermore, we determined that the agonistic VHH could support the proliferation of human-induced pluripotent stem cells (PSCs) and human mesenchymal stem cells, which are PSCs for regenerative medicine. In addition, the agonistic VHH could maintain the ability of mesenchymal stem cells to differentiate into adipocytes or osteocytes, indicating that it could maintain the properties of PSCs. These results suggest that the VHH agonist may function as an FGF2 mimetic in cell preparation of stem cells for regenerative medicine with better cost effectiveness.


Subject(s)
Fibroblast Growth Factor 2 , Protein Domains , Receptor, Fibroblast Growth Factor, Type 1 , Single-Domain Antibodies , Humans , Adipocytes/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Fibroblast Growth Factor 2/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Ligands , Mesoderm/cytology , Mesoderm/drug effects , Osteocytes/drug effects , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Regenerative Medicine , Signal Transduction/drug effects , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology
2.
J Biol Chem ; 299(1): 102729, 2023 01.
Article in English | MEDLINE | ID: mdl-36410439

ABSTRACT

Fibroblast growth factor receptor 1 (FGFR1) is a receptor tyrosine kinase that plays a major role in developmental processes and metabolism. The dysregulation of FGFR1 through genetic aberrations leads to skeletal and metabolic diseases as well as cancer. For this reason, FGFR1 is a promising therapeutic target, yet a very challenging one due to potential on-target toxicity. More puzzling is that both agonistic and antagonistic FGFR1 antibodies are reported to exhibit similar toxicity profiles in vivo, namely weight loss. In this study, we aimed to assess and compare the mechanism of action of these molecules to better understand this apparent contradiction. By systematically comparing the binding of these antibodies and the activation or the inhibition of the major FGFR1 signaling events, we demonstrated that the molecules displayed similar properties and can behave either as an agonist or antagonist depending on the presence or the absence of the endogenous ligand. We further demonstrated that these findings translated in xenografts mice models. In addition, using time-resolved FRET and mass spectrometry analysis, we showed a functionally distinct FGFR1 active conformation in the presence of an antibody that preferentially activates the FGFR substrate 2 (FRS2)-dependent signaling pathway, demonstrating that modulating the geometry of a FGFR1 dimer can effectively change the signaling outputs and ultimately the activity of the molecule in preclinical studies. Altogether, our results highlighted how bivalent antibodies can exhibit both agonistic and antagonistic activities and have implications for targeting other receptor tyrosine kinases with antibodies.


Subject(s)
Antibodies, Monoclonal , Receptor, Fibroblast Growth Factor, Type 1 , Signal Transduction , Animals , Humans , Mice , Neoplasms , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology
3.
Arch Pharm Res ; 43(12): 1311-1324, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33245516

ABSTRACT

Chronic inflammation in adipose tissue is the hallmark of obesity and a major risk factor for the development of obesity-induced insulin resistance. NLRP3 inflammasome regulates the maturation and secretion of pro-inflammatory cytokines, such as IL-1ß and IL-18, and was recently discovered to be involved in obesity-related metabolic diseases. Fibroblast growth factors (FGFs) such as FGF1, FGF10, and FGF21 are adipokines that regulate adipocyte development and metabolism, but reports on the effect of other FGFs on adipocytes are lacking. In the present study, the novel role of FGF2 in NLRP3 inflammasome activation was elucidated. Our results showed that FGF2 levels were increased during adipocyte differentiation and in the adipose tissue of high-fat diet (HFD)-induced obese mice. Recombinant FGF2 treatment upregulated inflammasome markers such as NLRP3, which was further exaggerated by TNF-ɑ treatment. Interestingly, ß-Klotho, a co-receptor of FGF21, was significantly decreased by FGF2 treatment. Results from mice confirmed the positive correlation between FGF2 and NLRP3 expression in epididymal and subcutaneous adipose tissue, while exercise training effectively reversed HFD-induced NLRP3 expression as well as FGF2 levels in both adipose depots. Our results suggest that FGF2 is an adipokine that may exacerbate the inflammatory response in adipocytes through NLRP3 inflammasome activation.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Fibroblast Growth Factor 2/pharmacology , Inflammasomes/metabolism , Inflammation/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obesity/metabolism , Subcutaneous Fat/drug effects , 3T3-L1 Cells , Adipocytes/immunology , Adipocytes/metabolism , Animals , Disease Models, Animal , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Inflammation/genetics , Inflammation/immunology , Klotho Proteins , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/immunology , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/agonists , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction , Subcutaneous Fat/immunology , Subcutaneous Fat/metabolism , Tumor Necrosis Factor-alpha/pharmacology
4.
Int J Mol Sci ; 21(7)2020 Mar 26.
Article in English | MEDLINE | ID: mdl-32225108

ABSTRACT

: Nonalcoholic steatohepatitis (NASH) is defined as a progressive form of nonalcoholic fatty liver disease (NAFLD) and is a common chronic liver disease that causes significant worldwide morbidity and mortality, and has no approved pharmacotherapy. Nevertheless, growing understanding of the molecular mechanisms underlying the development and progression of NASH has suggested multiple potential therapeutic targets and strategies to treat this disease. Here, we review this progress, with emphasis on the functional role of secretory proteins in the development and progression of NASH, in addition to the change of expression of various secretory proteins in mouse NASH models and human NASH subjects. We also highlight secretory protein-based therapeutic approaches that influence obesity-associated insulin resistance, liver steatosis, inflammation, and fibrosis, as well as the gut-liver and adipose-liver axes in the treatment of NASH.


Subject(s)
Glucagon-Like Peptide Receptors/agonists , Non-alcoholic Fatty Liver Disease/drug therapy , Receptor, Fibroblast Growth Factor, Type 1/agonists , Animals , Antioxidants/therapeutic use , Fibroblast Growth Factors/analogs & derivatives , Glucagon-Like Peptides/analogs & derivatives , Humans , Non-alcoholic Fatty Liver Disease/metabolism
5.
Neuropharmacology ; 170: 108070, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32229140

ABSTRACT

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


Subject(s)
Astrocytes/physiology , Fibroblast Growth Factor 2/physiology , Gamma Rhythm/physiology , Hippocampus/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Serotonin, 5-HT1A/physiology , Serotonin/physiology , Animals , Astrocytes/drug effects , Gamma Rhythm/drug effects , Hippocampus/drug effects , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/agonists , Serotonin 5-HT1 Receptor Agonists/pharmacology
6.
J Neural Transm (Vienna) ; 126(11): 1493-1500, 2019 11.
Article in English | MEDLINE | ID: mdl-31501979

ABSTRACT

In the present study, we developed an in vitro model of Huntington disease (HD) by transfecting primary rat hippocampal neurons with plasmids coding for m-htt exon 1 with different number of CAG repeats (18, 50 and 115) and demonstrated the influence of the length of polyQ sequence on neurite elongation. We found that exogenously applied FGF2 significantly rescued the m-htt-induced loss of neurite outgrowth. Moreover, the Enreptin peptide, an FGFR1 and NCAM dual agonist, had a similar neuritogenic effect to FGF2 in clinically relevant m-htt 50Q-expressing neurons. This study has developed an in vitro model of primary hippocampal neurons transfected with m-htt-coding vectors that is a powerful tool to study m-htt-related effects on neuronal placticity.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Hippocampus/drug effects , Huntingtin Protein/metabolism , Neural Cell Adhesion Molecules/pharmacology , Neuronal Outgrowth/drug effects , Neurons/drug effects , Oligopeptides/pharmacology , Receptor, Fibroblast Growth Factor, Type 1 , Animals , Disease Models, Animal , Huntingtin Protein/genetics , Neural Cell Adhesion Molecules/agonists , Rats , Receptor, Fibroblast Growth Factor, Type 1/agonists , Recombinant Proteins
7.
Neurobiol Aging ; 78: 155-165, 2019 06.
Article in English | MEDLINE | ID: mdl-30928883

ABSTRACT

Social isolation predominantly occurs in elderly people and it is strongly associated with cognitive decline. However, the mechanisms that produce isolation-related cognitive dysfunction during aging remain unclear. Here, we evaluated the cognitive, electrophysiological, and morphological effects of short- (4 weeks) and long-term (12 weeks) social isolation in aged male Wistar rats. Long-term but not short-term social isolation increased the plasma corticosterone levels and impaired spatial memory in the Morris water maze. Moreover, isolated animals displayed dampened hippocampal long-term potentiation in vivo, both in the dentate gyrus (DG) and CA1, as well as a specific reduction in the volume of the stratum oriens and spine density in CA1. Interestingly, social isolation induced a transient increase in hippocampal basic fibroblast growth factor (FGF2), whereas fibroblast growth factor receptor 1 (FGFR1) levels only increased after long-term isolation. Importantly, subchronic systemic administration of FGL, a synthetic peptide that activates FGFR1, rescued spatial memory in long-term isolated rats. These findings provide new insights into the neurobiological mechanisms underlying the detrimental effects on memory of chronic social isolation in the aged.


Subject(s)
Aging/psychology , Cognition , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Peptides/administration & dosage , Receptor, Fibroblast Growth Factor, Type 1/agonists , Social Isolation , Spatial Memory , Aging/metabolism , Animals , Cognitive Dysfunction/metabolism , Corticosterone/blood , Fibroblast Growth Factor 2/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Long-Term Potentiation , Male , Maze Learning , Peptides/pharmacology , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Time Factors
8.
Chem Commun (Camb) ; 55(18): 2672-2675, 2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30746545

ABSTRACT

Replacing expensive and thermally unstable growth factors with synthetic alternatives has been an important issue in stem cell-based regenerative medicines. Here we developed DNA aptamer-assemblies that act as functional mimics of basic fibroblast growth factor (bFGF), one of the essential factors for stem cell culture. The most potent aptamer assembly named TD0, composed solely of 76-mer single-stranded DNA, could support the self-renewal and pluripotency of induced pluripotent stem cells (iPSCs). This work presents the first application of DNA aptamer in the maintenance of iPSCs.


Subject(s)
Aptamers, Nucleotide/chemistry , Biocompatible Materials/chemistry , Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/pharmacology , Biocompatible Materials/metabolism , Biocompatible Materials/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/pharmacology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phosphorylation , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Up-Regulation/genetics
9.
Mol Metab ; 6(11): 1454-1467, 2017 11.
Article in English | MEDLINE | ID: mdl-29107292

ABSTRACT

OBJECTIVE: Fibroblast Growth Factor 21 (FGF21) is a potent stimulator of brown fat thermogenesis that improves insulin sensitivity, ameliorates hepatosteatosis, and induces weight loss by engaging the receptor complex comprised of Fibroblast Growth Factor Receptor 1 (FGFR1) and the requisite coreceptor ßKlotho. Previously, recombinant antibody proteins that activate the FGFR1/ßKlotho complex were proposed to act as an FGF21-mimetic; however, in vivo action of these engineered proteins has not been well studied. METHODS: We investigated the mechanism by which anti-FGFR1/ßKlotho bispecific antibody (bFKB1) stimulates thermogenesis in UCP1-expressing brown adipocytes using genetically engineered mice. Anti-FGFR1 agonist antibody was also used to achieve brown adipose tissue restricted activation in transgenic mice. RESULTS: Studies with global Ucp1-deficient mice and adipose-specific Fgfr1 deficient mice demonstrated that bFKB1 acts on targets distal to adipocytes and indirectly stimulates brown adipose thermogenesis in a UCP1-independent manner. Using a newly developed transgenic system, we also show that brown adipose tissue restricted activation of a transgenic FGFR1 expressed under the control of Ucp1 promoter does not stimulate energy expenditure. Finally, consistent with its action as a FGF21 mimetic, bFBK1 suppresses intake of saccharin-containing food and alcohol containing water in mice. CONCLUSIONS: Collectively, we propose that FGFR1/ßKlotho targeted therapy indeed mimics the action of FGF21 in vivo and stimulates UCP1-independent brown fat thermogenesis through receptors outside of adipocytes and likely in the nervous system.


Subject(s)
Membrane Proteins/immunology , Receptor, Fibroblast Growth Factor, Type 1/immunology , Thermogenesis/physiology , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Antibodies/metabolism , Energy Metabolism/physiology , Fibroblast Growth Factors/metabolism , Klotho Proteins , Membrane Proteins/agonists , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mitochondrial Proteins/metabolism , Obesity/metabolism , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Thermogenesis/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Weight Loss
10.
Cell Metab ; 26(5): 709-718.e3, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-28988823

ABSTRACT

Despite the different physiologic functions of FGF19 and FGF21 as hormonal regulators of fed and fasted metabolism, their pharmacologic administration causes similar increases in energy expenditure, weight loss, and enhanced insulin sensitivity in obese animals. Here, in genetic loss-of-function studies of the shared co-receptor ß-Klotho, we show that these pharmacologic effects are mediated through a common, tissue-specific pathway. Surprisingly, FGF19 and FGF21 actions in liver and adipose tissue are not required for their longer-term weight loss and glycemic effects. In contrast, ß-Klotho in neurons is essential for both FGF19 and FGF21 to cause weight loss and lower glucose and insulin levels. We further show an FGF21 mimetic antibody that activates the FGF receptor 1/ß-Klotho complex also requires neuronal ß-Klotho for its metabolic effects. These studies highlight the importance of the nervous system in mediating the beneficial weight loss and glycemic effects of endocrine FGF drugs.


Subject(s)
Antibodies, Monoclonal/pharmacology , Blood Glucose/drug effects , Fibroblast Growth Factors/pharmacology , Membrane Proteins/agonists , Nervous System/metabolism , Receptor, Fibroblast Growth Factor, Type 1/agonists , Weight Loss , Adipose Tissue/metabolism , Animals , Insulin Resistance , Klotho Proteins , Liver/metabolism , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Neurons/metabolism , Receptor, Fibroblast Growth Factor, Type 1/immunology
11.
J Nutr Biochem ; 49: 71-79, 2017 11.
Article in English | MEDLINE | ID: mdl-28886439

ABSTRACT

Excess carbohydrate intake causes obesity in humans. On the other hand, acute administration of fructose, glucose or sucrose in experimental animals has been shown to increase the plasma concentration of anti-obesity hormones such as glucagon-like peptide 1 (GLP-1) and Fibroblast growth factor 21 (FGF21), which contribute to reducing body weight. However, the secretion and action of GLP-1 and FGF21 in mice chronically fed a high-sucrose diet has not been investigated. To address the role of anti-obesity hormones in response to increased sucrose intake, we analyzed mice fed a high-sucrose diet, a high-starch diet or a normal diet for 15 weeks. Mice fed a high-sucrose diet showed resistance to body weight gain, in comparison with mice fed a high-starch diet or control diet, due to increased energy expenditure. Plasma FGF21 levels were highest among the three groups in mice fed a high-sucrose diet, whereas no significant difference in GLP-1 levels was observed. Expression levels of uncoupling protein 1 (UCP-1), FGF receptor 1c (FGFR1c) and ß-klotho (KLB) mRNA in brown adipose tissue were significantly increased in high sucrose-fed mice, suggesting increases in FGF21 sensitivity and energy expenditure. Expression of carbohydrate responsive element binding protein (ChREBP) mRNA in liver and brown adipose tissue was also increased in high sucrose-fed mice. These results indicate that FGF21 production in liver and brown adipose tissue is increased in high-sucrose diet and participates in resistance to weight gain.


Subject(s)
Adipose Tissue, Brown/metabolism , Diet, Carbohydrate Loading/adverse effects , Dietary Sucrose/adverse effects , Energy Metabolism , Fibroblast Growth Factors/agonists , Gene Expression Regulation, Developmental , Liver/metabolism , Adipose Tissue, Brown/growth & development , Adipose Tissue, White/growth & development , Adipose Tissue, White/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Insulin Resistance , Klotho Proteins , Liver/growth & development , Male , Membrane Proteins/agonists , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Nuclear Proteins/agonists , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Reproducibility of Results , Starch/adverse effects , Transcription Factors/agonists , Transcription Factors/genetics , Transcription Factors/metabolism , Uncoupling Protein 1/agonists , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Weight Gain
12.
Horm Mol Biol Clin Investig ; 30(2)2017 May 19.
Article in English | MEDLINE | ID: mdl-28525362

ABSTRACT

Fibroblast growth factor 21 (FGF21) analogs and FGF21 receptor agonists (FGF21RAs) that mimic FGF21 ligand activity constitute the new "FGF21-class" of anti-obesity and anti-diabetic molecules that improve insulin sensitivity, ameliorate hepatosteatosis and promote weight loss. The metabolic actions of FGF21-class proteins in obese mice are attributed to stimulation of brown fat thermogenesis and increased secretion of adiponectin. The therapeutic utility of this class of molecules is being actively investigated in clinical trials for the treatment of type 2 diabetes and non-alcoholic steatohepatitis (NASH). This review is focused on various FGF21-class molecules, their molecular designs and the preclinical and clinical activities. These molecules include modified FGF21 as well as agonistic antibodies against the receptor for FGF21, namely the complex of FGF receptor 1 (FGFR1) and the obligatory coreceptor ßKlotho (KLB). In addition, a novel approach to increase endogenous FGF21 activity by inhibiting the FGF21-degrading protease fibroblast activation protein (FAP) is discussed.


Subject(s)
Fibroblast Growth Factors/metabolism , Obesity/metabolism , Receptor, Fibroblast Growth Factor, Type 1/agonists , Animals , Energy Metabolism/drug effects , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/therapeutic use , Gene Expression Regulation/drug effects , Humans , Obesity/drug therapy , Obesity/genetics , Protein Stability , Signal Transduction/drug effects , Transcription, Genetic
13.
Biochem Biophys Res Commun ; 480(1): 55-60, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27717820

ABSTRACT

Fibroblast growth factor receptor-1c (FGFR1c)/ßKlotho (KLB) complex is a receptor of fibroblast growth factor 21 (FGF21). Pharmacologically, FGF21 shows anti-obesity and anti-diabetic effects upon peripheral administration. Here, we report the development of an artificial peptide agonist to the FGFR1c/KLB heterodimer complex. The peptide, F91-8A07 (LPGRTCREYPDLWWVRCY), was discovered from random peptide T7 phage display and selectively bound to the FGFR1c/KLB complex, but not to FGFR1c and KLB individually. After subsequent peptide dimerization using a short polyethyleneglycol (PEG) linker, the dimeric F91-8A07 peptide showed higher potent agonist activity than that of FGF21 in cultured primary human adipocytes. Moreover, the dimeric peptide led to an expression of the early growth response protein-1 (Egr-1) mRNA in vivo, which is a target gene of FGFR1c. To the best of our knowledge, this is the first report of a FGFR1c/KLB complex-selective artificial peptide agonist.


Subject(s)
Cell Surface Display Techniques , Membrane Proteins/agonists , Peptides/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/agonists , Adipocytes/drug effects , Animals , Bacteriophage T7 , Dimerization , Drug Discovery , Fibroblast Growth Factors/chemistry , Humans , Klotho Proteins , Male , Membrane Proteins/metabolism , Mice, Inbred BALB C , Multiprotein Complexes/metabolism , Peptide Library , Peptides/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism
14.
FEBS Lett ; 590(1): 53-67, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26762170

ABSTRACT

Mechanisms underlying the association between fibroblastic growth factor 23 (FGF-23) and inflammation are uncertain. We found that FGF-23 was markedly up-regulated in LPS/INF-γ-induced proinflammatory M1 macrophages and Hyp mouse-derived peritoneal macrophages, but not in IL-4-induced M2 anti-inflammatory macrophages. NF-КB and JAK/STAT1 pathways mediated the increased transcription of FGF-23 in response to M1 polarization. FGF-23 stimulated TNF-α, but not IL-6, expression in M0 macrophages and suppressed Arginase-1 expression in M2 macrophages through FGFR-mediated mechanisms. 1,25(OH)2 D stimulated Arginase-1 expression and inhibited FGF-23 stimulation of TNF-α. FGF-23 has proinflammatory paracrine functions and counter-regulatory actions to 1,25(OH)2 D on innate immune responses.


Subject(s)
Calcitriol/metabolism , Fibroblast Growth Factors/metabolism , Macrophages/metabolism , Paracrine Communication , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptors, Calcitriol/agonists , Signal Transduction , Animals , Arginase/antagonists & inhibitors , Arginase/chemistry , Arginase/genetics , Arginase/metabolism , Cells, Cultured , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/genetics , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , HEK293 Cells , Humans , Immunity, Innate/drug effects , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Paracrine Communication/drug effects , Promoter Regions, Genetic/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RAW 264.7 Cells , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Calcitriol/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction/drug effects
15.
PLoS One ; 10(8): e0135577, 2015.
Article in English | MEDLINE | ID: mdl-26263555

ABSTRACT

Latent transforming growth factor-beta-1 binding protein-2 (LTBP-2) belongs to the fibrillin-LTBP superfamily of extracellular matrix proteins. LTBPs and fibrillins are involved in the sequestration and storage of latent growth factors, particularly transforming growth factor ß (TGF-ß), in tissues. Unlike other LTBPs, LTBP-2 does not covalently bind TGF-ß, and its molecular functions remain unclear. We are screening LTBP-2 for binding to other growth factors and have found very strong saturable binding to fibroblast growth factor-2 (FGF-2) (Kd = 1.1 nM). Using a series of recombinant LTBP-2 fragments a single binding site for FGF-2 was identified in a central region of LTBP-2 consisting of six tandem epidermal growth factor-like (EGF-like) motifs (EGFs 9-14). This region was also shown to contain a heparin/heparan sulphate-binding site. FGF-2 stimulation of fibroblast proliferation was completely negated by the addition of 5-fold molar excess of LTBP-2 to the assay. Confocal microscopy showed strong co-localisation of LTBP-2 and FGF-2 in fibrotic keloid tissue suggesting that the two proteins may interact in vivo. Overall the study indicates that LTBP-2 is a potent inhibitor of FGF-2 that may influence FGF-2 bioactivity during wound repair particularly in fibrotic tissues.


Subject(s)
Binding Sites , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/chemistry , Latent TGF-beta Binding Proteins/chemistry , Latent TGF-beta Binding Proteins/metabolism , Cell Line , Cell Proliferation/drug effects , Fibrillins , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/metabolism , Heparin/metabolism , Humans , Keloid/metabolism , Latent TGF-beta Binding Proteins/pharmacology , Microfilament Proteins/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recombinant Proteins , Skin/metabolism
16.
EBioMedicine ; 2(7): 730-43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26288846

ABSTRACT

Dissipating excess calories as heat through therapeutic stimulation of brown adipose tissues (BAT) has been proposed as a potential treatment for obesity-linked disorders. Here, we describe the generation of a humanized effector-less bispecific antibody that activates fibroblast growth factor receptor (FGFR) 1/ßKlotho complex, a common receptor for FGF21 and FGF19. Using this molecule, we show that antibody-mediated activation of FGFR1/ßKlotho complex in mice induces sustained energy expenditure in BAT, browning of white adipose tissue, weight loss, and improvements in obesity-associated metabolic derangements including insulin resistance, hyperglycemia, dyslipidemia and hepatosteatosis. In mice and cynomolgus monkeys, FGFR1/ßKlotho activation increased serum high-molecular-weight adiponectin, which appears to contribute over time by enhancing the amplitude of the metabolic benefits. At the same time, insulin sensitization by FGFR1/ßKlotho activation occurs even before the onset of weight loss in a manner that is independent of adiponectin. Together, selective activation of FGFR1/ßKlotho complex with a long acting therapeutic antibody represents an attractive approach for the treatment of type 2 diabetes and other obesity-linked disorders through enhanced energy expenditure, insulin sensitization and induction of high-molecular-weight adiponectin.


Subject(s)
Adipose Tissue, Brown/metabolism , Antibodies, Bispecific/pharmacology , Insulin/pharmacology , Membrane Proteins/agonists , Receptor, Fibroblast Growth Factor, Type 1/agonists , Adiponectin/metabolism , Adipose Tissue, Brown/drug effects , Animals , Cell Line , Energy Metabolism/drug effects , Fibroblast Growth Factors/pharmacology , HEK293 Cells , Humans , Klotho Proteins , Macaca fascicularis , Male , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Obese , Protein Binding/drug effects , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Thermogenesis/drug effects , Weight Loss/drug effects
17.
PLoS Pathog ; 10(6): e1004165, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24967908

ABSTRACT

Host arginase 1 (arg1) expression is a significant contributor to the pathogenesis of progressive visceral leishmaniasis (VL), a neglected tropical disease caused by the intracellular protozoan Leishmania donovani. Previously we found that parasite-induced arg1 expression in macrophages was dependent on STAT6 activation. Arg1 expression was amplified by, but did not require, IL-4, and required de novo synthesis of unknown protein(s). To further explore the mechanisms involved in arg1 regulation in VL, we screened a panel of kinase inhibitors and found that inhibitors of growth factor signaling reduced arg1 expression in splenic macrophages from hamsters with VL. Analysis of growth factors and their signaling pathways revealed that the Fibroblast Growth Factor Receptor 1 (FGFR-1) and Insulin-like Growth Factor 1 Receptor (IGF-1R) and a number of downstream signaling proteins were activated in splenic macrophages isolated from hamsters infected with L. donovani. Recombinant FGF-2 and IGF-1 increased the expression of arg1 in L. donovani infected hamster macrophages, and this induction was augmented by IL-4. Inhibition of FGFR-1 and IGF-1R decreased arg1 expression and restricted L. donovani replication in both in vitro and ex vivo models of infection. Inhibition of the downstream signaling molecules JAK and AKT also reduced the expression of arg1 in infected macrophages. STAT6 was activated in infected macrophages exposed to either FGF-2 or IGF-1, and STAT6 was critical to the FGFR-1- and IGF-1R-mediated expression of arg1. The converse was also true as inhibition of FGFR-1 and IGF-1R reduced the activation of STAT6 in infected macrophages. Collectively, these data indicate that the FGFR/IGF-1R and IL-4 signaling pathways converge at STAT6 to promote pathologic arg1 expression and intracellular parasite survival in VL. Targeted interruption of these pathological processes offers an approach to restrain this relentlessly progressive disease.


Subject(s)
Arginase/metabolism , Leishmaniasis, Visceral/immunology , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, IGF Type 1/agonists , STAT6 Transcription Factor/metabolism , Signal Transduction , Th2 Cells/immunology , Animals , Arginase/genetics , Cell Line , Cells, Cultured , Disease Progression , Enzyme Induction/drug effects , Host-Parasite Interactions/drug effects , Interleukin-4/metabolism , Leishmania donovani/growth & development , Leishmania donovani/immunology , Leishmania donovani/pathogenicity , Leishmania donovani/physiology , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/physiopathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/parasitology , Mesocricetus , Protein Kinase Inhibitors/pharmacology , RNA Interference , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , STAT6 Transcription Factor/agonists , STAT6 Transcription Factor/antagonists & inhibitors , STAT6 Transcription Factor/genetics , Signal Transduction/drug effects , Th2 Cells/drug effects , Th2 Cells/metabolism , Th2 Cells/parasitology
18.
Biochem Biophys Res Commun ; 441(2): 387-92, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24157794

ABSTRACT

New findings show that neurotrophic and antidepressant effects of 5-HT in brain can, in part, be mediated by activation of the 5-HT1A receptor protomer in the hippocampal and raphe FGFR1-5-HT1A heteroreceptor complexes enhancing the FGFR1 signaling. The dynamic agonist modulation of the FGFR1-5-HT1A heteroreceptor complexes and their recruitment of ß-arrestin is now determined in cellular models with focus on its impact on 5-HT1AR and FGFR1 homodimerization in the heteroreceptor complexes based on BRET(2) assays. The findings show that coagonist treatment with 8-OH-DPAT and FGF2 but not treatment with the 5-HT1A agonist alone markedly increases the BRETmax values and significantly reduces the BRET50 values of 5HT1A homodimerization. The effects of FGF2 or FGF20 with or without the 5-HT1A agonist were also studied on the FGFR1 homodimerization of the heteroreceptor complexes. FGF2 produced a marked and rapid increase in FGFR1 homodimerization which partially declined over a 10min period. Cotreatment with FGF2 and 5-HT1A agonist blocked this decline in FGFR1 homodimerization. Furthermore, FGF2 alone produced a small increase in the BRET(2) signal from the 5-HT1A-ß-arrestin2 receptor-protein complex which was additive to the marked effect of 8-OH-DPAT alone. Taken together, the participation of 5-HT1A and FGFR1 homodimers and recruitment of ß-arrestin2 was demonstrated in the FGFR1-5-HT1A heteroreceptor complexes upon agonist treatments.


Subject(s)
8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Arrestins/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin Receptor Agonists/pharmacology , Fibroblast Growth Factor 2/pharmacology , HEK293 Cells , Humans , Protein Conformation , Protein Multimerization , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Serotonin, 5-HT1A/chemistry , Serotonin 5-HT1 Receptor Agonists/pharmacology , beta-Arrestins
19.
Sci Transl Med ; 4(162): 162ra153, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197570

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a distinctive member of the FGF family with potent beneficial effects on lipid, body weight, and glucose metabolism and has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to native FGF21, we have developed a monoclonal antibody, mimAb1, that binds to ßKlotho with high affinity and specifically activates signaling from the ßKlotho/FGFR1c (FGF receptor 1c) receptor complex. In obese cynomolgus monkeys, injection of mimAb1 led to FGF21-like metabolic effects, including decreases in body weight, plasma insulin, triglycerides, and glucose during tolerance testing. Mice with adipose-selective FGFR1 knockout were refractory to FGF21-induced improvements in glucose metabolism and body weight. These results in obese monkeys (with mimAb1) and in FGFR1 knockout mice (with FGF21) demonstrated the essential role of FGFR1c in FGF21 function and suggest fat as a critical target tissue for the cytokine and antibody. Because mimAb1 depends on ßKlotho to activate FGFR1c, it is not expected to induce side effects caused by activating FGFR1c alone. The unexpected finding of an antibody that can activate FGF21-like signaling through cell surface receptors provided preclinical validation for an innovative therapeutic approach to diabetes and obesity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus/drug therapy , Fibroblast Growth Factors/immunology , Glucuronidase/metabolism , Obesity/drug therapy , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Antibodies, Monoclonal/pharmacology , Body Weight/genetics , Diabetes Mellitus/blood , Epitopes/chemistry , Glucose/metabolism , Glucuronidase/immunology , Humans , Klotho Proteins , Macaca fascicularis , Mice , Obesity/blood , Obesity/complications , Phosphates/blood , Receptor, Fibroblast Growth Factor, Type 1/agonists , Signal Transduction/drug effects , Triglycerides/blood
20.
J Neurochem ; 114(1): 74-86, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20374425

ABSTRACT

Basic fibroblast growth factor (FGF2, bFGF) is the most extensively studied member of the FGF family and is involved in neurogenesis, differentiation, neuroprotection, and synaptic plasticity in the CNS. FGF2 executes its pleiotropic biologic actions by binding, dimerizing, and activating FGF receptors (FGFRs). The present study reports the physiologic impact of various FGF2-FGFR1 contact sites employing three different synthetic peptides, termed canofins, designed based on structural analysis of the interactions between FGF2 and FGFR1. Canofins mimic the cognate ligand interaction with the receptor and preserve the neuritogenic and neuroprotective properties of FGF2. Canofins were shown by surface plasmon resonance analysis to bind to FGFR1 and promote receptor activation. However, FGF2-induced receptor phosphorylation was inhibited by canofins, indicating that canofins are partial FGFR agonists. Furthermore, canofins were demonstrated to induce neuronal differentiation determined by neurite outgrowth from cerebellar granule neurons, and this effect was dependent on FGFR activation. Additionally, canofins acted as neuroprotectants, promoting survival of cerebellar granule neurons induced to undergo apoptosis. Our results suggest that canofins mirror the effect of specific interaction sites in FGF2 for FGFR. Thus, canofins are valuable pharmacological tools to study the functional roles of specific molecular interactions of FGF2 with FGFR.


Subject(s)
Dendrimers/pharmacology , Fibroblast Growth Factor 2/metabolism , Neurons/drug effects , Oligopeptides/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebellum/cytology , Dendrimers/chemistry , Drug Partial Agonism , Fibroblast Growth Factor 2/chemistry , Ligands , Models, Molecular , Neurites/drug effects , Neurites/physiology , Neurons/cytology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oligopeptides/chemistry , Phosphorylation , Rats , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...