Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 337
Filter
1.
Biochimie ; 225: 26-39, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38740172

ABSTRACT

Fibroblast growth factor 21 (FGF21) is pivotal in regulating energy metabolism, highlighting substantial therapeutic potential for non-alcoholic steatohepatitis (NASH). Previously, we reported a long-acting FGF21 fusion protein, PsTag-FGF21, which was prepared by genetically fusing human FGF21 with a 648-residue polypeptide (PsTag). While this fusion protein demonstrated therapeutic efficacy against NASH, our final product analysis revealed the presence of fixed impurities resistant to effective removal, indicating potential degradation of PsTag-FGF21. Here, we enriched and analyzed the impurities, confirming our hypothesis regarding the C-terminal degradation of PsTag-FGF21. We now describe a new variant developed to eliminate the C-terminal degradation. By introducing one mutation located at the C-terminal of PsTag-FGF21(V169L), we demonstrated that the new molecule, PsTag-FGF21(V169L), exhibits many improved attributes. Compared with PsTag-FGF21, PsTag-FGF21(V169L) displayed elevated bioactivity and stability, along with a twofold enhanced binding affinity to the coreceptor ß-Klotho. In vivo, the circulating half-life of PsTag-FGF21(V169L) was further enhanced compared with that of PsTag-FGF21. In NASH mice, PsTag-FGF21(V169L) demonstrated efficacy with sustained improvements in multiple metabolic parameters. Besides, PsTag-FGF21(V169L) demonstrated the ability to alleviate NASH by decreasing hepatocyte apoptosis. The superior biophysical, pharmacokinetic, and pharmacodynamic properties, along with the positive metabolic effects, imply that further clinical development of PsTag-FGF21(V169L) as a metabolic therapy for NASH patients may be warranted.


Subject(s)
Fibroblast Growth Factors , Non-alcoholic Fatty Liver Disease , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/chemistry , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Mice , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Male , Klotho Proteins , Mice, Inbred C57BL , Protein Stability
2.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(3): 269-274, 2024 Mar 15.
Article in Chinese | MEDLINE | ID: mdl-38557379

ABSTRACT

OBJECTIVES: To observe the correlation between growth impairment induced by long-term oral glucocorticoids (GC) therapy and the ratio of FGF23/Klotho in children with primary nephrotic syndrome (PNS). METHODS: A prospective study was conducted on 56 children with GC-sensitive PNS who had discontinued GC therapy for more than 3 months and revisited the Department of Pediatrics of the First Affiliated Hospital of Henan University of Traditional Chinese Medicine between June 2022 and December 2022. After monitoring qualitative and quantitative urine protein levels upon admission, the children with proteinuria relapse were treated with GC (GC group; n=29), while those without relapse did not receive GC treatment (non-GC group; n=27). In addition, 29 healthy children aged 3 to prepuberty were selected as the control group. Height, bone age, growth rate, and the FGF23/Klotho ratio were compared among the groups. The correlations of the FGF23/Klotho ratio with height, bone age, and growth rate were analyzed. RESULTS: The FGF23/Klotho ratio in the GC group was significantly higher than that in the non-GC group after 1 month of GC therapy (P<0.05), and the height and bone age growth rates within 6 months were lower than those in the non-GC group (P<0.05). Correlation analysis showed significant negative correlations between the FGF23/Klotho ratio after 1 month of treatment and the growth rates of height and bone age within 6 months in children with PNS (r=-0.356 and -0.436, respectively; P<0.05). CONCLUSIONS: The disturbance in FGF23/Klotho homeostasis is one of the mechanisms underlying the growth impairment caused by long-term oral GC therapy.


Subject(s)
Fibroblast Growth Factor-23 , Glucocorticoids , Glucuronidase , Growth Disorders , Klotho Proteins , Child , Humans , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/drug effects , Glucocorticoids/adverse effects , Prospective Studies , Recurrence , Klotho Proteins/chemistry , Klotho Proteins/drug effects , Fibroblast Growth Factor-23/chemistry , Fibroblast Growth Factor-23/drug effects , Growth Disorders/chemically induced
3.
Trends Pharmacol Sci ; 44(12): 862-864, 2023 12.
Article in English | MEDLINE | ID: mdl-37778940

ABSTRACT

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.


Subject(s)
Fibroblast Growth Factors , Klotho Proteins , Humans , Fibroblast Growth Factors/chemistry , Glucuronidase/metabolism , Signal Transduction/physiology , Receptors, Fibroblast Growth Factor/metabolism
4.
Toxicol Appl Pharmacol ; 428: 115673, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34364948

ABSTRACT

Pegbelfermin (PGBF) is a PEGylated fibroblast growth factor 21 (FGF21) analogue in development for treatment of nonalcoholic steatohepatitis (NASH). Mouse models highlight potential utility of FGF21 in NASH, but also suggest negative effects on bone, though these findings are confounded by profound FGF21-related decreases in body mass/growth. This study aimed to profile PGBF-related bone effects in adult nonhuman primates after long-term, clinically-relevant exposures. Adult male cynomolgus monkeys received weekly subcutaneous PGBF (0.3, 0.75 mg/kg) or control injections for 1 year (n = 5/group). Assessments included body weight, clinical chemistry, adiponectin levels, bone turnover biomarkers, skeletal radiography, pharmacokinetics, immunogenicity, and histopathology. Bone densitometry and body composition were evaluated in vivo and/or ex vivo with dual-energy x-ray absorptiometry, peripheral quantitative computed tomography, and biomechanical strength testing. After 1 year of PGBF administration, there was clear evidence of sustained PGBF pharmacology in monkeys (peak increase in serum adiponectin of 1.7× and 2.35× pretest at 0.3 and 0.75 mg/kg PGBF, respectively) and decreased body weight compared with control at exposures comparable to those tested in humans. At 0.75 mg/kg PGBF, pharmacologically-mediated reductions in lean mass, lean area, and fat area were observed relative to controls. There were no PGBF-related effects on bone biomarkers, radiography, densitometry, or strength. Together, these data demonstrate that PGBF did not adversely alter bone metabolism, density, or strength following 1 year of dosing at clinically relevant (0.7-2.2× human AUC[0-168 h] at 20 mg once weekly), pharmacologically-active exposures in adult monkeys, suggesting a low potential for negative effects on bone quality in adult humans.


Subject(s)
Bone Density/drug effects , Bone Remodeling/drug effects , Fibroblast Growth Factors/analogs & derivatives , Polyethylene Glycols/administration & dosage , Animals , Bone Density/physiology , Bone Remodeling/physiology , Drug Administration Schedule , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/chemistry , Haplorhini , Macaca fascicularis , Male , Polyethylene Glycols/chemistry , Time Factors
5.
FASEB J ; 35(2): e21286, 2021 02.
Article in English | MEDLINE | ID: mdl-33484478

ABSTRACT

Human Fibroblast Growth Factor 19 (FGF19) and mouse ortholog Fgf15 play similar roles in liver regeneration and metabolism via the activation of Fgfr4/b-klotho (Klb). Monomeric FGF19 and dimeric Fgf15 are both necessary for liver regeneration and proper bile acid (BA) metabolism. FGF19 elicits stronger effects than Fgf15 on glucose and fatty acid metabolism and only FGF19 induces hepatocellular carcinoma (HCC). However, inhibiting FGF19/FGFR4 signaling in HCC patients is associated with toxicity due to elevated BA levels. Here, we examine the structure/function relationship in Fgf15/FGF19 to better understand the molecular basis for their distinct functions. We demonstrate that FGF19 is a more effective activator of Fgfr4 and of downstream signaling (Erk, Plcg1) than Fgf15. Furthermore, we use site-directed mutagenesis to show that the presence or absence of an unpaired cysteine in Fgf15/19 modulates ligand structure and determines the ability of these molecules to induce hepatocyte proliferation, with monomers being more potent activators. Consistent with these findings, an engineered dimeric variant of FGF19 is less effective than wild-type FGF19 at inducing liver growth in cooperation with the Wnt-enhancer RSPO3. In contrast to effects on proliferation, monomeric and dimeric ligands equally inhibited the expression of Cyp7a1, the enzyme catalyzing the rate limiting step in BA production. Thus, structure and function of Fgf15/FGF19 are intricately linked, explaining why FGF19, but not Fgf15, induces liver tumorigenesis. Our data provide insight into FGF19/FGFR4 signaling and may inform strategies to target this pathway while limiting on-target toxicity due to dysregulation of BA production or induction of hepatocyte proliferation.


Subject(s)
Cell Proliferation , Fibroblast Growth Factors/metabolism , Hepatocytes/metabolism , Protein Multimerization , Signal Transduction , Amino Acid Motifs , Animals , Cholesterol 7-alpha-Hydroxylase/metabolism , Female , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , HEK293 Cells , Humans , Male , Mice , Mutation , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Thrombospondins/metabolism
6.
Proc Natl Acad Sci U S A ; 117(46): 29025-29034, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33144503

ABSTRACT

As a physiological regulator of bile acid homeostasis, FGF19 is also a potent insulin sensitizer capable of normalizing plasma glucose concentration, improving lipid profile, ameliorating fatty liver disease, and causing weight loss in both diabetic and diet-induced obesity mice. There is therefore a major interest in developing FGF19 as a therapeutic agent for treating type 2 diabetes and cholestatic liver disease. However, the known tumorigenic risk associated with prolonged FGF19 administration is a major hurdle in realizing its clinical potential. Here, we show that nonmitogenic FGF19 variants that retain the full beneficial glucose-lowering and bile acid regulatory activities of WT FGF19 (FGF19WT) can be engineered by diminishing FGF19's ability to induce dimerization of its cognate FGF receptors (FGFR). As proof of principle, we generated three such variants, each with a partial defect in binding affinity to FGFR (FGF19ΔFGFR) and its coreceptors, i.e., ßklotho (FGF19ΔKLB) or heparan sulfate (FGF19ΔHBS). Pharmacological assays in WT and db/db mice confirmed that these variants incur a dramatic loss in mitogenic activity, yet are indistinguishable from FGF19WT in eliciting glycemic control and regulating bile acid synthesis. This approach provides a robust framework for the development of safer and more efficacious FGF19 analogs.


Subject(s)
Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Mitogens/metabolism , Animals , Bile Acids and Salts/metabolism , Diabetes Mellitus, Type 2 , Dimerization , Disease Models, Animal , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/pharmacology , Genetic Engineering , Glucose/metabolism , Hep G2 Cells , Homeostasis , Humans , Klotho Proteins , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Obese/genetics , Receptors, Fibroblast Growth Factor/metabolism
7.
J Clin Endocrinol Metab ; 105(12)2020 12 01.
Article in English | MEDLINE | ID: mdl-32951052

ABSTRACT

CONTEXT: Higher fibroblast growth factor-23 (FGF23) concentrations are associated with heart failure and mortality in diverse populations, but the strengths of associations differ markedly depending up on which assay is used. OBJECTIVE: We sought to evaluate whether iron deficiency, inflammation, or kidney function account for differences in the strengths of associations between these 2 FGF23 assays with clinical outcomes. DESIGN: Case cohort study from the Cardiovascular Health Study. SETTING: A total of 844 community-dwelling individuals aged 65 years or older with and without chronic kidney disease were followed for 10 years. OUTCOMES: Outcomes included death, incident heart failure (HF), and incident myocardial infarction (MI). Exposure was baseline intact and C-terminal FGF23. Using modified Cox models, adjusting sequentially we tested whether observed associations of each assay with outcomes were attenuated by iron status, inflammation, kidney function, or their combinations. RESULTS: FGF23 measured by either assay was associated with mortality in unadjusted analysis (intact FGF23 hazard ratio [HR] per 2-fold higher 1.45; 95% CI, 1.25-1.68; C-terminal FGF23 HR 1.38; 95% CI, 1.26-1.50). Adjustment for kidney function completely attenuated associations of intact FGF23 with mortality (HR 1.00; 95% CI, 0.85-1.17), but had much less influence on the association of C-terminal FGF23, for which results remained significant after adjustment (HR 1.15; 95% CI, 1.04-1.28). Attenuation was much less with adjustment for iron status or inflammation. Results were similar for the HF end point. Neither C-terminal or intact FGF23 was associated with MI risk. CONCLUSIONS: The relationship of FGF23 with clinical end points is markedly different depending on the type of FGF23 assay used. The associations of biologically active FGF23 with clinical end points may be confounded by kidney disease, and thus much weaker than previously thought.


Subject(s)
Anemia, Iron-Deficiency/diagnosis , Fibroblast Growth Factors/blood , Inflammation/diagnosis , Kidney/physiology , Renal Insufficiency, Chronic/diagnosis , Aged , Aged, 80 and over , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/epidemiology , Case-Control Studies , Cohort Studies , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/analysis , Fibroblast Growth Factors/chemistry , Heart Failure/blood , Heart Failure/diagnosis , Heart Failure/epidemiology , Heart Failure/etiology , Humans , Incidence , Inflammation/blood , Inflammation/complications , Inflammation/epidemiology , Iron Deficiencies , Kidney Function Tests , Male , Prognosis , Protein Domains , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/epidemiology , Risk Factors
8.
Physiol Rep ; 8(14): e14505, 2020 07.
Article in English | MEDLINE | ID: mdl-32671946

ABSTRACT

The voltage-gated sodium (Nav) channel complex is comprised of pore-forming α subunits (Nav1.1-1.9) and accessory regulatory proteins such as the intracellular fibroblast growth factor 14 (FGF14). The cytosolic Nav1.6 C-terminal tail binds directly to FGF14 and this interaction modifies Nav1.6-mediated currents with effects on intrinsic excitability in the brain. Previous studies have identified the FGF14V160 residue within the FGF14 core domain as a hotspot for the FGF14:Nav1.6 complex formation. Here, we used three short amino acid peptides around FGF14V160 to probe for the FGF14 interaction with the Nav1.6 C-terminal tail and to evaluate the activity of the peptide on Nav1.6-mediated currents. In silico docking predicts FLPK to bind to FGF14V160 with the expectation of interfering with the FGF14:Nav1.6 complex formation, a phenotype that was confirmed by the split-luciferase assay (LCA) and surface plasmon resonance (SPR), respectively. Whole-cell patch-clamp electrophysiology studies demonstrate that FLPK is able to prevent previously reported FGF14-dependent phenotypes of Nav1.6 currents, but that its activity requires the FGF14 N-terminal tail, a domain that has been shown to contribute to Nav1.6 inactivation independently from the FGF14 core domain. In medium spiny neurons in the nucleus accumbens, where both FGF14 and Nav1.6 are abundantly expressed, FLPK significantly increased firing frequency by a mechanism consistent with the ability of the tetrapeptide to interfere with Nav1.6 inactivation and potentiate persistent Na+ currents. Taken together, these results indicate that FLPK might serve as a probe for characterizing molecular determinants of neuronal excitability and a peptide scaffold to develop allosteric modulators of Nav channels.


Subject(s)
Fibroblast Growth Factors/metabolism , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Neurons/metabolism , Peptide Fragments/pharmacology , Recombinant Fusion Proteins/metabolism , Animals , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , NAV1.6 Voltage-Gated Sodium Channel/chemistry , NAV1.6 Voltage-Gated Sodium Channel/genetics , Neurons/drug effects , Peptide Fragments/chemistry , Protein Binding , Protein Interaction Maps , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification
9.
Molecules ; 25(15)2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32722255

ABSTRACT

Disruption of protein:protein interactions (PPIs) that regulate the function of voltage-gated Na+ (Nav) channels leads to neural circuitry aberrations that have been implicated in numerous channelopathies. One example of this pathophysiology is mediated by dysfunction of the PPI between Nav1.6 and its regulatory protein fibroblast growth factor 14 (FGF14). Thus, peptides derived from FGF14 might exert modulatory actions on the FGF14:Nav1.6 complex that are functionally relevant. The tetrapeptide Glu-Tyr-Tyr-Val (EYYV) mimics surface residues of FGF14 at the ß8-ß9 loop, a structural region previously implicated in its binding to Nav1.6. Here, peptidomimetics derived from EYYV (6) were designed, synthesized, and pharmacologically evaluated to develop probes with improved potency. Addition of hydrophobic protective groups to 6 and truncation to a tripeptide (12) produced a potent inhibitor of FGF14:Nav1.6 complex assembly. Conversely, addition of hydrophobic protective groups to 6 followed by addition of an N-terminal benzoyl substituent (19) produced a potentiator of FGF14:Nav1.6 complex assembly. Subsequent functional evaluation using whole-cell patch-clamp electrophysiology confirmed their inverse activities, with 12 and 19 reducing and increasing Nav1.6-mediated transient current densities, respectively. Overall, we have identified a negative and positive allosteric modulator of Nav1.6, both of which could serve as scaffolds for the development of target-selective neurotherapeutics.


Subject(s)
Fibroblast Growth Factors/chemistry , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Peptidomimetics/chemical synthesis , Peptidomimetics/pharmacology , Allosteric Regulation , Binding Sites , Drug Design , Fibroblast Growth Factors/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Docking Simulation , Molecular Structure , NAV1.6 Voltage-Gated Sodium Channel/chemistry , Peptidomimetics/chemistry , Protein Binding/drug effects , Protein Structure, Secondary
10.
Biochim Biophys Acta Mol Cell Res ; 1867(10): 118786, 2020 10.
Article in English | MEDLINE | ID: mdl-32599005

ABSTRACT

BACKGROUND: Protein interactions between voltage-gated sodium (Nav) channels and accessory proteins play an essential role in neuronal firing and plasticity. However, a surprisingly limited number of kinases have been identified as regulators of these molecular complexes. We hypothesized that numerous as-of-yet unidentified kinases indirectly regulate the Nav channel via modulation of the intracellular fibroblast growth factor 14 (FGF14), an accessory protein with numerous unexplored phosphomotifs and required for channel function in neurons. METHODS: Here we present results from an in-cell high-throughput screening (HTS) against the FGF14: Nav1.6 complex using >3000 diverse compounds targeting an extensive range of signaling pathways. Regulation by top kinase targets was then explored using in vitro phosphorylation, biophysics, mass-spectrometry and patch-clamp electrophysiology. RESULTS: Compounds targeting Janus kinase 2 (JAK2) were over-represented among HTS hits. Phosphomotif scans supported by mass spectrometry revealed FGF14Y158, a site previously shown to mediate both FGF14 homodimerization and interactions with Nav1.6, as a JAK2 phosphorylation site. Following inhibition of JAK2, FGF14 homodimerization increased in a manner directly inverse to FGF14:Nav1.6 complex formation, but not in the presence of the FGF14Y158A mutant. Patch-clamp electrophysiology revealed that through Y158, JAK2 controls FGF14-dependent modulation of Nav1.6 channels. In hippocampal CA1 pyramidal neurons, the JAK2 inhibitor Fedratinib reduced firing by a mechanism that is dependent upon expression of FGF14. CONCLUSIONS: These studies point toward a novel mechanism by which levels of JAK2 in neurons could directly influence firing and plasticity by controlling the FGF14 dimerization equilibrium, and thereby the availability of monomeric species for interaction with Nav1.6.


Subject(s)
Fibroblast Growth Factors/metabolism , Janus Kinase 2/metabolism , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Action Potentials/drug effects , Amino Acid Sequence , Animals , Fibroblast Growth Factors/chemistry , HEK293 Cells , High-Throughput Screening Assays , Hippocampus/cytology , Humans , Mice, Inbred C57BL , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Kinase Inhibitors/analysis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Multimerization/drug effects , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Reproducibility of Results , src-Family Kinases/metabolism
11.
EBioMedicine ; 54: 102719, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32259714

ABSTRACT

BACKGROUND: Disruption of bile acid (BA) homeostasis plays a key role in intestinal inflammation. The gut-liver axis is the main site for the regulation of BA synthesis and BA pool size via the combined action of the nuclear Farnesoid X Receptor (FXR) and the enterokine Fibroblast Growth Factor 19 (FGF19). Increasing evidence have linked derangement of BA metabolism with dysbiosis and mucosal inflammation. Thus, here we aimed to investigate the potential action of an FGF19 analogue on intestinal microbiota and inflammation. METHODS: A novel engineered non-tumorigenic variant of the FGF19 protein, M52-WO 2016/0168219 was generated. WT and FXRnull mice were injected with AAV-FGF19-M52 or the control AAV-GFP and subjected to Sodium Dextran Sulphate-induced colitis. FINDINGS: FGF19-M52 reduced BA synthesis and pool size, modulated its composition and protected mice from intestinal inflammation. These events were coupled with preservation of the intestinal epithelial barrier integrity, inhibition of inflammatory immune response and modulation of microbiota composition. Interestingly, FGF19-M52-driven systemic and local anti-inflammatory activity was completely abolished in Farnesoid X Receptor (FXR)null mice, thus underscoring the need of FXR to guarantee enterocytes' fitness and complement FGF19 anti-inflammatory activity. To provide a translational perspective, we also show that circulating FGF19 levels are reduced in patients with Crohn's disease. INTERPRETATION: Reactivation of the FXR-FGF19 axis in a murine model of intestinal inflammation could bona fide provide positive changes in BA metabolism with consequent reduction of intestinal inflammation and modulation of microbiota. These results point to the therapeutic potential of FGF19 in the treatment of intestinal inflammation with concomitant derangement of BA homeostasis. FUNDING: A. Moschetta is funded by MIUR-PRIN 2017 <- 2017J3E2W2; Italian Association for Cancer Research (AIRC, IG 23239); Interreg V-A Greece-Italy 2014-2020-SILVER WELLBEING, MIS5003627; HDHL-INTIMIC EuJPI-FATMAL; MIUR PON "R&I" 2014-2020-ARS01_01220. No money has been paid by NGM Biopharmaceuticals or any other agency to write this article.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/microbiology , Crohn Disease/microbiology , Fibroblast Growth Factors/metabolism , Gastrointestinal Microbiome , Peptides/therapeutic use , Animals , Bile Acids and Salts/metabolism , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Crohn Disease/drug therapy , Crohn Disease/metabolism , Female , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Peptides/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use
12.
Cancer Sci ; 111(5): 1750-1760, 2020 May.
Article in English | MEDLINE | ID: mdl-32061104

ABSTRACT

Hepatocellular carcinoma (HCC) is a common and particularly fatal form of cancer for which very few drugs are effective. The fibroblast growth factor 19 (FGF19) has been viewed as a driver of HCC development and a potential Ab target for developing novel HCC therapy. However, a previously developed anti-FGF19 Ab disrupted FGF19's normal regulatory function and caused severe bile-acid-related side-effects despite of having potent antitumor effects in preclinical models. Here, we developed novel human Abs (G1A8 and HS29) that specifically target the N-terminus of FGF19. Both Abs inhibited FGF19-induced HCC cell proliferation in vitro and significantly suppressed HCC tumor growth in mouse models. Importantly, no bile-acid-related side effects were observed in preclinical cynomolgus monkeys. Fundamentally, our study demonstrates that it is possible to target FGF19 for anti-HCC therapies without adversely affecting its normal bile acid regulatory function, and highlights the exciting promise of G1A8 or HS29 as potential therapy for HCC.


Subject(s)
Antibodies/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Fibroblast Growth Factors/immunology , Liver Neoplasms/drug therapy , Animals , Antibodies/chemistry , Antibodies/immunology , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Bile Acids and Salts/blood , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Disease Models, Animal , Epitopes , Female , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Macaca fascicularis , Male , Mice
13.
Nat Chem Biol ; 16(3): 351-360, 2020 03.
Article in English | MEDLINE | ID: mdl-31932717

ABSTRACT

Polypeptide GalNAc-transferase T3 (GalNAc-T3) regulates fibroblast growth factor 23 (FGF23) by O-glycosylating Thr178 in a furin proprotein processing motif RHT178R↓S. FGF23 regulates phosphate homeostasis and deficiency in GALNT3 or FGF23 results in hyperphosphatemia and familial tumoral calcinosis. We explored the molecular mechanism for GalNAc-T3 glycosylation of FGF23 using engineered cell models and biophysical studies including kinetics, molecular dynamics and X-ray crystallography of GalNAc-T3 complexed to glycopeptide substrates. GalNAc-T3 uses a lectin domain mediated mechanism to glycosylate Thr178 requiring previous glycosylation at Thr171. Notably, Thr178 is a poor substrate site with limiting glycosylation due to substrate clashes leading to destabilization of the catalytic domain flexible loop. We suggest GalNAc-T3 specificity for FGF23 and its ability to control circulating levels of intact FGF23 is achieved by FGF23 being a poor substrate. GalNAc-T3's structure further reveals the molecular bases for reported disease-causing mutations. Our findings provide an insight into how GalNAc-T isoenzymes achieve isoenzyme-specific nonredundant functions.


Subject(s)
Fibroblast Growth Factors/chemistry , N-Acetylgalactosaminyltransferases/metabolism , Animals , CHO Cells , Cricetulus , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Glycopeptides/chemistry , Glycosylation , Humans , Isoenzymes/metabolism , Lectins/metabolism , N-Acetylgalactosaminyltransferases/physiology , Threonine/metabolism , Polypeptide N-acetylgalactosaminyltransferase
14.
Eur J Pharmacol ; 871: 172920, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-31935396

ABSTRACT

Stable electrical activity in cardiac myocytes is the basis of maintaining normal myocardial systolic and diastolic function. Cardiac ionic currents and their associated regulatory proteins are crucial to myocyte excitability and heart function. Fibroblast growth factor homologous factors (FHFs) are intracellular noncanonical fibroblast growth factors (FGFs) that are incapable of activating FGF receptors. The main functions of FHFs are to regulate ion channels and influence excitability, which are processes involved in sustaining normal cardiac function. In addition to their regulatory effect on ion channels, FHFs can be regulators of cardiac hypertrophic signaling and alter signaling pathways, including the protein kinase, NFB, and p53 pathways, which are related to the pathological processes of heart diseases. This review emphasizes FHF-mediated regulation of cardiac excitability and the association of FHFs with cardiac arrhythmias and explores the idea that abnormal FHFs may be an unrecognized cause of cardiac disorders.


Subject(s)
Arrhythmias, Cardiac/metabolism , Fibroblast Growth Factors/metabolism , Ion Channels/metabolism , Animals , Arrhythmias, Cardiac/pathology , Fibroblast Growth Factors/chemistry , Humans , Myocytes, Cardiac/metabolism
15.
Mol Pharm ; 17(1): 284-300, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31794223

ABSTRACT

Therapeutic protein depots have limited clinical success because of the presence of critical preparation barriers such as low encapsulation, uncontrolled release, and activity loss during processing and storage. In the present study, we used our novel protein-nanoencapsulation (into sugar-glass nanoparticle; SGnP) platform to prepare a protein depot to overcome the abovementioned formidable challenges. The SGnP-mediated microparticle protein depot has been validated using four model proteins (bovine serum albumin, horseradish peroxidase, fibroblastic growth factor, and epidermal growth factor) and model biodegradable poly(lactic-co-glycolic acid) polymer system. The results show that our protein-nanoencapsulation-mediated platform provides a new generic platform to prepare a protein depot through the conventional emulsion method of any polymer and single/multiple protein systems. This protein depot has the required pharmaceutical properties such as high encapsulation efficiency, burst-free sustained release, and protein preservation during processing and storage, making it suitable for off-the-shelf use in therapeutic protein delivery and tissue engineering applications.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Proteins/administration & dosage , Delayed-Action Preparations/chemistry , Drug Compounding/methods , Emulsions , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/metabolism , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Glass/chemistry , Horseradish Peroxidase/chemistry , Horseradish Peroxidase/metabolism , Humans , MCF-7 Cells , Microscopy, Electron, Scanning , Nanoparticles/ultrastructure , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Proteins/chemistry , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Spheroids, Cellular/drug effects , Sugars
16.
J Biomed Mater Res B Appl Biomater ; 108(2): 326-332, 2020 02.
Article in English | MEDLINE | ID: mdl-31016841

ABSTRACT

Several nerve conduits have been investigated for their potential as alternative sources of autografts for bridging neural gaps. However, autologous nerve transplants remain the most effective for nerve repair. We examined clinically approved nerve conduits containing collagen and polyglycolic acid (PGA-c) combined with collagen-binding basic fibroblast growth factor (bFGF) containing a polycystic kidney disease (PKD) domain and collagen binding domain (CBD) (bFGF-PKD-CBD) in a rat 15-mm sciatic nerve critical-size defect model. The treatment groups were: PGA-c immersed in phosphate-buffered saline (PBS) (PGA-c/PBS group), bFGF (PGA-c/bFGF group), or bFGF-PKD-CBD (PGA-c/bFGF-PKD-CBD group), and no treatment (Defect group). Gait and histological analyses were performed. Four weeks after treatment, the recovery rate of the paw print area was significantly greater in the PGA-c/bFGFPKD-CBD group than the PGA-c/PBS and PGA-c/bFGF groups. Mean intensity of paw prints was significantly greater in the PGA-c/bFGF-PKD-CBD group than the PGA-c/PBS and Defect groups. Swing time was significantly greater in the PGA-c/PBS, PGA-c/bFGF, and PGA-c/bFGF-PKD-CBD groups than the Defect group. At 8 weeks, all three parameters were significantly greater in the PGA-c/PBS, PGA-c/bFGF, and PGA-c/bFGF-PKD-CBD groups than the Defect group. Regenerated myelinated fibers were observed in 7/8 (87.5%) rats in the PGA-c/bFGF-PKD-CBD group after 8 weeks, and in 1/8 (12.5%) and 3/8 (37.5%) rats in the PGA-c/PBS and PGA-c/bFGF groups, respectively. PGA-c/bFGF-PKD-CBD composites may be promising biomaterials for promoting functional recovery of long-distance peripheral nerve defects in clinical practice.


Subject(s)
Biocompatible Materials/chemistry , Collagen/chemistry , Fibroblast Growth Factors/metabolism , Gait/physiology , Polyglycolic Acid/chemistry , Sciatic Nerve/metabolism , Tissue Scaffolds/chemistry , Animals , Autografts/metabolism , Behavior, Animal , Cell Proliferation , Collagen/metabolism , Disease Models, Animal , Fibroblast Growth Factors/chemistry , Humans , Male , Nerve Regeneration/physiology , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/therapy , Polyglycolic Acid/metabolism , Protein Binding , Protein Domains , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tissue Engineering
17.
Handb Exp Pharmacol ; 262: 281-308, 2020.
Article in English | MEDLINE | ID: mdl-31792685

ABSTRACT

FGF23 is a phosphotropic hormone produced by the bone. FGF23 works by binding to the FGF receptor-Klotho complex. Klotho is expressed in several limited tissues including the kidney and parathyroid glands. This tissue-restricted expression of Klotho is believed to determine the target organs of FGF23. FGF23 reduces serum phosphate by suppressing the expression of type 2a and 2c sodium-phosphate cotransporters in renal proximal tubules. FGF23 also decreases 1,25-dihydroxyvitamin D levels by regulating the expression of vitamin D-metabolizing enzymes, which results in reduced intestinal phosphate absorption. Excessive actions of FGF23 cause several types of hypophosphatemic rickets/osteomalacia characterized by impaired mineralization of bone matrix. In contrast, deficient actions of FGF23 result in hyperphosphatemic tumoral calcinosis with high 1,25-dihydroxyvitamin D levels. These results indicate that FGF23 is a physiological regulator of phosphate and vitamin D metabolism and indispensable for the maintenance of serum phosphate levels.


Subject(s)
Hyperphosphatemia , Hypophosphatemia , Osteomalacia , Bone and Bones , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Humans , Minerals/chemistry , Phosphates/chemistry , Phosphates/metabolism
18.
Sci Rep ; 9(1): 16890, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31729429

ABSTRACT

Multiple voltage-gated Na+ (Nav) channelopathies can be ascribed to subtle changes in the Nav macromolecular complex. Fibroblast growth factor 14 (FGF14) is a functionally relevant component of the Nav1.6 channel complex, a causative link to spinocerebellar ataxia 27 (SCA27) and an emerging risk factor for neuropsychiatric disorders. Yet, how this protein:channel complex is regulated in the cell is still poorly understood. To search for key cellular pathways upstream of the FGF14:Nav1.6 complex, we have developed, miniaturized and optimized an in-cell assay in 384-well plates by stably reconstituting the FGF14:Nav1.6 complex using the split-luciferase complementation assay. We then conducted a high-throughput screening (HTS) of 267 FDA-approved compounds targeting known mediators of cellular signaling. Of the 65 hits initially detected, 24 were excluded based on counter-screening and cellular toxicity. Based on target analysis, potency and dose-response relationships, 5 compounds were subsequently repurchased for validation and confirmed as hits. Among those, the tyrosine kinase inhibitor lestaurtinib was highest ranked, exhibiting submicromolar inhibition of FGF14:Nav1.6 assembly. While providing evidence for a robust in-cell HTS platform that can be adapted to search for any channelopathy-associated regulatory proteins, these results lay the potential groundwork for repurposing cancer drugs for neuropsychopharmacology.


Subject(s)
Antineoplastic Agents , High-Throughput Screening Assays/methods , Protein Interaction Maps/physiology , Voltage-Gated Sodium Channel Agonists/isolation & purification , Voltage-Gated Sodium Channel Blockers/isolation & purification , Voltage-Gated Sodium Channels/drug effects , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical/methods , Fibroblast Growth Factors/agonists , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/chemistry , HEK293 Cells , Humans , Multiprotein Complexes/agonists , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/chemistry , NAV1.6 Voltage-Gated Sodium Channel/drug effects , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Protein Binding , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/metabolism
19.
EBioMedicine ; 48: 462-477, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31631034

ABSTRACT

BACKGROUND: The development of a clinically useful fibroblast growth factor 21 (FGF21) hormone has been impeded by its inherent instability and weak FGF receptor (FGFR) binding affinity. There is an urgent need for innovative approaches to overcome these limitations. METHODS: We devised a structure-based chimerisation strategy in which we substituted the thermally labile and low receptor affinity core of FGF21 with an HS binding deficient endocrinised core derived from a stable and high receptor affinity paracrine FGF1 (FGF1ΔHBS). The thermal stability, receptor binding ability, heparan sulfate and ßKlotho coreceptor dependency of the chimera were measured using a thermal shift assay, SPR, SEC-MALS and cell-based studies. The half-life, tissue distribution, glucose lowering activity and adipose tissue remodeling were analyzed in normal and diabetic mice and monkeys. FINDINGS: The melting temperature of the engineered chimera (FGF1ΔHBS-FGF21C-tail) increased by ∼22 °C relative to wild-type FGF21 (FGF21WT), and resulted in a ∼5-fold increase in half-life in vivo. The chimera also acquired an ability to bind the FGFR1c isoform - the principal receptor that mediates the metabolic actions of FGF21 - and consequently was dramatically more effective than FGF21WT in correcting hyperglycemia and in ameliorating insulin resistance in db/db mice. Our chimeric FGF21 also exerted a significant beneficial effect on glycemic control in spontaneous diabetic cynomolgus monkeys. INTERPRETATION: Our study describes a structure-based chimerisation approach that effectively mitigates both the intrinsically weak receptor binding affinities and short half-lives of endocrine FGFs, and advance the development of the FGF21 hormone into a potentially useful drug for Type 2 diabetes.


Subject(s)
Fibroblast Growth Factors/metabolism , Metabolic Diseases/metabolism , Paracrine Communication , Adipocytes/metabolism , Animals , Biomarkers , Chromatography, High Pressure Liquid , Disease Models, Animal , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Gene Expression , Humans , Insulin/metabolism , Male , Metabolic Diseases/drug therapy , Metabolic Diseases/etiology , Mice , Models, Molecular , Paracrine Communication/drug effects , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recombinant Proteins , Structure-Activity Relationship
20.
Int J Mol Sci ; 20(20)2019 Oct 11.
Article in English | MEDLINE | ID: mdl-31614494

ABSTRACT

We investigated the effects of a fibrin-hyaluronic acid hydrogel (FBG-HA) and fibroblast growth factor 18 (FGF-18) for nucleus pulposus (NP) regeneration. Healthy bovine (n = 4) and human degenerated NP cells (n = 4) were cultured for 14 days in FBG-HA hydrogel with FGF-18 (∆51-mutant or wild-type) in the culture medium. Gene expression, DNA content, and glycosaminoglycan (GAG) synthesis were evaluated on day 7 and 14. Additionally, histology was performed. Human NP cells cultured in FBG-HA hydrogel showed an increase in collagen type II (COL2) and carbonic anhydrase XII (CA12) gene expression after 14 or 7 days of culture, respectively. GAG release into the conditioned medium increased over 14 days. Healthy bovine NP cells showed increased gene expression of ACAN from day 7 to day 14. Wild type FGF-18 up-regulated CA12 gene expression of human NP cells. Histology revealed an increase of proteoglycan deposition upon FGF-18 stimulation in bovine but not in human NP cells. The FBG-HA hydrogel had a positive modulatory effect on human degenerated NP cells. Under the tested conditions, no significant effect of FGF-18 was observed on cell proliferation or GAG synthesis in human NP cells.


Subject(s)
Cell Culture Techniques/methods , Fibroblast Growth Factors/pharmacology , Hyaluronic Acid/chemistry , Nucleus Pulposus/cytology , Animals , Biomimetics , Carbonic Anhydrases/genetics , Cattle , Cells, Cultured , Collagen Type II/metabolism , Fibroblast Growth Factors/chemistry , Glycosaminoglycans/metabolism , Humans , Hyaluronic Acid/pharmacology , Hydrogels/chemistry , Nucleus Pulposus/drug effects , Nucleus Pulposus/metabolism , Phenotype , Regeneration
SELECTION OF CITATIONS
SEARCH DETAIL