Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
BMC Biotechnol ; 23(1): 43, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37789318

RESUMEN

BACKGROUND: The major safety concern of the clinical application of wild type FGF19 (FGF19WT) emerges given that its extended treatment causes hepatocellular carcinoma. Therefore, we previously generated a safer FGF19 variant - FGF19ΔKLB, which have same effects on glycemic control and bile acid production but much less mitogenic activity. However, it remains unclear as to whether FGF19ΔKLB ameliorates intrahepatic cholestasis. RESULTS: We found that, similar to that of FGF19WT, the chronic administration of FGF19ΔKLB protects mice from cholestatic liver injury in these two models. The therapeutic benefits of FGF19ΔKLB on cholestatic liver damage are attributable, according to the following mechanistic investigation, to the reduction of BA production, liver inflammation, and fibrosis. More importantly, FGF19ΔKLB did not induce any tumorigenesis effects during its prolonged treatment. CONCLUSIONS: Together, our findings raise hope that FGF19ΔKLB may represent a useful therapeutic strategy for the treatment of intrahepatic cholestasis.


Asunto(s)
Colestasis Intrahepática , Colestasis , Animales , Ratones , Ácidos y Sales Biliares , Colestasis/tratamiento farmacológico , Colestasis/patología , Colestasis Intrahepática/tratamiento farmacológico , Colestasis Intrahepática/genética , Colestasis Intrahepática/patología , Modelos Animales de Enfermedad , Hígado
2.
Proc Natl Acad Sci U S A ; 117(46): 29025-29034, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33144503

RESUMEN

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.


Asunto(s)
Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Mitógenos/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Diabetes Mellitus Tipo 2 , Dimerización , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/química , Factores de Crecimiento de Fibroblastos/farmacología , Ingeniería Genética , Glucosa/metabolismo , Células Hep G2 , Homeostasis , Humanos , Proteínas Klotho , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Obesos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
3.
BMC Med Genet ; 20(1): 33, 2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30777021

RESUMEN

BACKGROUND: Genome-wide association studies (GWASs) of a large cohort of subjects with chronic obstructive pulmonary disease (COPD) have successfully identified multiple risk genes, including fibroblast growth factor 7 (FGF7). However, the underlying molecular mechanism influencing function of FGF7 and risk of COPD remains further study. METHODS: In this study, we replicated the genetic association of variants near the FGF7 gene in 258 Chinese Han patients with COPD and 311 healthy controls. Additionally, we functionally evaluated a candidate causal variant upstream of the FGF7 gene. RESULTS: The most significant association was observed at rs12905203 (P = 5.9 × 10- 3, odd ratio, OR = 1.516) that explains associations of previously reported variants at the FGF7 locus. Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation-quantitative polymerase chain reaction (ChIP-qPCR) assays showed that the risk allele of the variant was bound to activator protein 1 transcription factors (c-Fos and c-Jun) with a significantly reduced affinity and associated with decreased mRNA expression of FGF7 in fibroblast cells at both resting and PMA/Ionomycin-stimulated conditions. Overexpression of c-Fos and c-Jun proteins or stimulation with PMA/Ionomycin significantly increases mRNA expression of FGF7 in fibroblast cells. Bioinformatic analysis showed that the variant overlaps with multiple genetic regulatory marks, suggesting the regulatory DNA element might function as an enhancer for the FGF7 gene. Luciferase enhancer activity assays demonstrated that the DNA sequences carrying the variant produce enhancer activity while the risk allele of the variant reduces its activity. CONCLUSIONS: In this study, we demonstrated a consistent association of the FGF7 gene with COPD and mechanistically characterized a candidate functional variant upstream of the FGF7 gene. These data highlighted the important role of the risk variant and the FGF7 gene in influencing risk for COPD.


Asunto(s)
Factor 7 de Crecimiento de Fibroblastos/genética , Estudio de Asociación del Genoma Completo/métodos , Polimorfismo de Nucleótido Simple , Enfermedad Pulmonar Obstructiva Crónica/genética , Factor de Transcripción AP-1/metabolismo , Anciano , Estudios de Casos y Controles , China/etnología , Femenino , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/etnología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo
4.
BMC Biotechnol ; 18(1): 33, 2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-29843712

RESUMEN

BACKGROUND: High level of serum fibroblast growth factor 23 (FGF23) is implicated in the development and progression of diabetic nephropathy (DN), making it a crucial factor in the pathogenesis of DN. FGF23 is also tightly correlated with inflammation in the progression of DN. The aim of this study was to explore whether the C-terminal of FGF23 (FGF23C-tail), an antagonist that can block the FGF23 signaling pathway by competing with intact FGF23, could exhibit a therapeutic effect on DN. RESULTS: Biochemical data and histological examination showed that FGF23 C-tail administration ameliorated the functional and morphological abnormalities of db/db mice with DN without changing the levels of circulating FGF23 and phosphate. Evaluation of morphology and fibrosis by Masson's trichrome staining and IHC staining of fibronectin, PCR, and western blot analysis showed that FGF23C-tail prevents diabetes-induced fibrosis in db/db mice. Importantly, FGF23C-tail decreased the levels of inflammatory cytokines in serum and renal tissues. CONCLUSION: FGF23C-tail may improve diabetic nephropathy by decreasing inflammation and fibrosis in db/db mice, suggesting that blocking of FGF23 action remains an important therapeutic target for the prevention or attenuation of the progression of DN.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/uso terapéutico , Inflamación/tratamiento farmacológico , Animales , Citocinas/análisis , Citocinas/sangre , Citocinas/orina , Nefropatías Diabéticas/patología , Factor-23 de Crecimiento de Fibroblastos , Fibrosis , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
5.
BMC Biotechnol ; 17(1): 58, 2017 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-28676059

RESUMEN

BACKGROUND: Fibroblast growth factor 21 (FGF21) is an endocrine-acting hormone that has the potential to treat diabetic nephropathy. However, development of FGF21 into a therapeutic has been hindered due to its low intrinsic bio-stability. In our previous study, we have developed a recombinant human FGF21 (rhFGF21) variant by site-directed mutagenesis and solid-phase PEGylation, which retained its biological function. The aim of this study is to elucidate whether the therapeutic effect of PEGylated rhFGF21 (PEG-rhFGF21) on diabetic nephropathy in DIO (diet induced obesity) mice is more significant than rhFGF21 in vivo. RESULTS: After administration with rhFGF21 and PEG-rhFGF21 for 2 months, biochemical data and histological examination showed that PEG-rhFGF21 significantly lowered lipid levels in the kidney, decreased urine albumin/creatinine ratio (ACR) and improved mesangial expansion, demonstrating that PEG-rhFGF21 was more efficacious in ameliorating functional and morphological abnormalities induced by diabetic nephropathy in db/db and DIO mice. CONCLUSIONS: Our findings suggest that PEG-rhFGF21 treatment is more effective in treating diabetic nephropathy than rhFGF21, through enhancements of systemic metabolic alterations and anti-inflammatory mechanisms. These findings help provide a theoretical basis to develop more long-acting and efficacious protein drugs for diabetic nephropathy.


Asunto(s)
Preparaciones de Acción Retardada/administración & dosificación , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/patología , Factores de Crecimiento de Fibroblastos/administración & dosificación , Obesidad/tratamiento farmacológico , Proteínas Recombinantes/administración & dosificación , Animales , Nefropatías Diabéticas/etiología , Relación Dosis-Respuesta a Droga , Factores de Crecimiento de Fibroblastos/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/complicaciones , Obesidad/patología , Proteínas Recombinantes/genética , Resultado del Tratamiento
6.
Bioconjug Chem ; 25(1): 63-71, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24341776

RESUMEN

Production of protein therapeutics often involves in vitro refolding from bacterial inclusion bodies and subsequent PEGylation to improve protein stability and plasma half-life. Here, we devised a novel strategy for one-step production of site-specific mono-PEGylated proteins with good bioactivity and improved biostability by integrating PEGylation and protein refolding (IPPR). Using lysozyme and recombinant human fibroblast growth factor 21 (rhFGF21) as model proteins, we showed that both PEGylation and refolding of denatured proteins have been simultaneously accomplished by IPPR with high efficiency of refolding yield and bioconjugation. PEGylated rhFGF21 by IPPR has a similar capacity as the native rhFGF21 to stimulate glucose uptake in 3T3-L1 cells, but exhibits prolonged blood glucose and triglyceride lowering activity levels in the ob/ob diabetic mouse model. Hence, IPPR will significantly facilitate the generation of protein therapeutics.


Asunto(s)
Factores de Crecimiento de Fibroblastos/química , Muramidasa/química , Polietilenglicoles/química , Replegamiento Proteico , Células 3T3-L1 , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Ratones , Ratones Obesos , Muramidasa/metabolismo , Polietilenglicoles/metabolismo , Polietilenglicoles/farmacocinética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
7.
Biochem Pharmacol ; 221: 116039, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38301966

RESUMEN

Translocator protein (18 kDa) (TSPO) plays an important role in retinal neuroinflammation in the early stage of diabetic retinopathy (DR). Studies have found that a FGF1 variant (FGF1ΔHBS) with reduced proliferative potency exerts excellent anti-inflammatory effects and potential therapeutic value for diabetic complications. In this study, intravitreal injection of FGF1ΔHBS was administrated every week for one month in db/db mice, which are genetically predisposed to develop type 2 diabetes mellitus and early retinopathy. Changes in retinal function and structure in the animal models were detected by electrophysiology (ERG) and optical tomography coherence (OCT). TSPO expression and retinal inflammation were analyzed by immunofluorescence, Western blot and real-time qPCR. In the retina of T2D (db/db) mice, FGF1 was significantly down-regulated while FGFR1 was up-regulated (both p < 0.05). TSPO and retinal inflammatory factors were all up-regulated. TSPO and FGFR1 were mainly co-stained in the inner retina. After FGF1ΔHBS treatment, ERG showed that the total amplitude of dark-adapted b-wave and oscillating potentials (Ops) was significantly improved, and OCT showed that the thickness of the retina around the optical nerve head was significantly preserved in T2D mice (all p < 0.05). The TSPO signal was significantly suppressed by FGF1ΔHBS. The activation of NF-κB p65 and the expression of inflammatory factors such as TNF-α, IL-1ß, IL-6, COX-2, MIP-1α, and iNOS were all significantly down-regulated (all p < 0.05). Collectively, our current data demonstrated that intravitreal FGF1ΔHBS treatment can effectively inhibit retinal inflammation via suppressing TSPO signal and to preserve retinal function and structure in a T2D mouse model.


Asunto(s)
Diabetes Mellitus Tipo 2 , Retinopatía Diabética , Ratones , Animales , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 1 de Crecimiento de Fibroblastos/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Retina/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Proteínas Portadoras/metabolismo
8.
J Vis Exp ; (196)2023 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-37458432

RESUMEN

Several models have been developed to investigate angiogenesis in vivo. However, most of these models are complex and expensive, require specialized equipment, or are hard to perform for subsequent quantitative analysis. Here we present a modified matrix gel plug assay to evaluate angiogenesis in vivo. In this protocol, vascular cells were mixed with matrix gel in the presence or absence of pro-angiogenic or anti-angiogenic reagents, and then subcutaneously injected into the back of recipient mice. After 7 days, phosphate buffer saline containing dextran-FITC is injected via the tail vein and circulated in vessels for 30 min. Matrix gel plugs are collected and embedded with tissue embedding gel, then 12 µm sections are cut for fluorescence detection without staining. In this assay, dextran-FITC with high molecular weight (~150,000 Da) can be used to indicate functional vessels for detecting their length, while dextran-FITC with low molecular weight (~4,400 Da) can be used to indicate the permeability of neo-vessels. In conclusion, this protocol can provide a reliable and convenient method for the quantitative study of angiogenesis in vivo.


Asunto(s)
Dextranos , Neovascularización Fisiológica , Ratones , Animales , Fluoresceína-5-Isotiocianato , Fenómenos Fisiológicos Cardiovasculares
9.
Biomaterials ; 301: 122276, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37579564

RESUMEN

Photoimmunotherapy has been acknowledged to be an unprecedented strategy to obtain significantly improved cancer treatment efficacy. In this regard, the exploitation of high-performance multimodal phototheranostic agents is highly desired. Apart from tailoring electron donors, acceptor engineering is gradually rising as a deliberate approach in this field. Herein, we rationally designed a family of aggregation-induced emission (AIE)-active compounds with the same donors but different acceptors based on the acceptor engineering. Through finely adjusting the functional groups on electron acceptors, the electron affinity of electron acceptors and the conformation of the compounds were simultaneously modulated. It was found that one of the molecules (named DCTIC), bearing a moderately electrophilic electron acceptor and the best planarity, exhibited optimal phototheranostic properties in terms of light-harvesting ability, fluorescence emission, reactive oxygen species (ROS) production, and photothermal performance. For the purpose of amplified therapeutic outcomes, DCTIC was fabricated into tumor and mitochondria dual-targeted DCTIC nanoparticles (NPs), which afforded good performance in the fluorescence/photoacoustic/photothermal trimodal imaging-guided photodynamic/photothermal-synergized cancer immunotherapy with the combination of programmed cell death protein-1 (PD-1) antibody. Not only the primary tumors were totally eradicated, but efficient growth inhibition of distant tumors was also realized.


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Fototerapia , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Inmunoterapia , Mitocondrias , Nanomedicina Teranóstica , Oxidantes , Imagen Multimodal , Línea Celular Tumoral
10.
Cell Metab ; 35(6): 1022-1037.e6, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37167965

RESUMEN

Type 2 diabetes (T2D) is a major health and economic burden worldwide. Despite the availability of multiple drugs for short-term management, sustained remission of T2D is currently not achievable pharmacologically. Intracerebroventricular administration of fibroblast growth factor 1 (icvFGF1) induces sustained remission in T2D rodents, propelling intense research efforts to understand its mechanism of action. Whether other FGFs possess similar therapeutic benefits is currently unknown. Here, we show that icvFGF4 also elicits a sustained antidiabetic effect in both male db/db mice and diet-induced obese mice by activating FGF receptor 1 (FGFR1) expressed in glucose-sensing neurons within the mediobasal hypothalamus. Specifically, FGF4 excites glucose-excited (GE) neurons while inhibiting glucose-inhibited (GI) neurons. Moreover, icvFGF4 restores the percentage of GI neurons in db/db mice. Importantly, intranasal delivery of FGF4 alleviates hyperglycemia in db/db mice, paving the way for non-invasive therapy. We conclude that icvFGF4 holds significant therapeutic potential for achieving sustained remission of T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hiperglucemia , Ratones , Animales , Masculino , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Factor 4 de Crecimiento de Fibroblastos/uso terapéutico , Roedores/metabolismo , Glucosa/metabolismo , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Factores de Crecimiento de Fibroblastos/uso terapéutico , Factores de Crecimiento de Fibroblastos/metabolismo
11.
Redox Biol ; 49: 102219, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34990928

RESUMEN

A cumulative and progressively developing cardiomyopathy induced by adriamycin (ADR)-based chemotherapy is a major obstacle for its clinical application. However, there is a lack of safe and effective method to protect against ADR-induced cardiotoxicity. Here, we found that mRNA and protein levels of FGF1 were decreased in ADR-treated mice, primary cardiomyocytes and H9c2 cells, suggesting the potential effect of FGF1 to protect against ADR-induced cardiotoxicity. Then, we showed that treatment with a FGF1 variant (FGF1ΔHBS) with reduced proliferative potency significantly prevented ADR-induced cardiac dysfunction as well as ADR-associated cardiac inflammation, fibrosis, and hypertrophy. The mechanistic study revealed that apoptosis and oxidative stress, the two vital pathological factors in ADR-induced cardiotoxicity, were largely alleviated by FGF1ΔHBS treatment. Furthermore, the inhibitory effects of FGF1ΔHBS on ADR-induced apoptosis and oxidative stress were regulated by decreasing p53 activity through upregulation of Sirt1-mediated p53 deacetylation and enhancement of murine double minute 2 (MDM2)-mediated p53 ubiquitination. Upregulation of p53 expression or cardiac specific-Sirt1 knockout (Sirt1-CKO) almost completely abolished FGF1ΔHBS-induced protective effects in cardiomyocytes. Based on these findings, we suggest that FGF1ΔHBS may be a potential therapeutic agent against ADR-induced cardiotoxicity.


Asunto(s)
Cardiotoxicidad , Doxorrubicina , Animales , Apoptosis , Cardiotoxicidad/patología , Doxorrubicina/efectos adversos , Factor 1 de Crecimiento de Fibroblastos/genética , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Ratones , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
12.
Nat Commun ; 12(1): 7256, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34907199

RESUMEN

Several members of the FGF family have been identified as potential regulators of glucose homeostasis. We previously reported that a low threshold of FGF-induced FGF receptor 1c (FGFR1c) dimerization and activity is sufficient to evoke a glucose lowering activity. We therefore reasoned that ligand identity may not matter, and that besides paracrine FGF1 and endocrine FGF21, other cognate paracrine FGFs of FGFR1c might possess such activity. Indeed, via a side-by-side testing of multiple cognate FGFs of FGFR1c in diabetic mice we identified the paracrine FGF4 as a potent anti-hyperglycemic FGF. Importantly, we found that like FGF1, the paracrine FGF4 is also more efficacious than endocrine FGF21 in lowering blood glucose. We show that paracrine FGF4 and FGF1 exert their superior glycemic control by targeting skeletal muscle, which expresses copious FGFR1c but lacks ß-klotho (KLB), an obligatory FGF21 co-receptor. Mechanistically, both FGF4 and FGF1 upregulate GLUT4 cell surface abundance in skeletal muscle in an AMPKα-dependent but insulin-independent manner. Chronic treatment with rFGF4 improves insulin resistance and suppresses adipose macrophage infiltration and inflammation. Notably, unlike FGF1 (a pan-FGFR ligand), FGF4, which has more restricted FGFR1c binding specificity, has no apparent effect on food intake. The potent anti-hyperglycemic and anti-inflammatory properties of FGF4 testify to its promising potential for use in the treatment of T2D and related metabolic disorders.


Asunto(s)
Factor 4 de Crecimiento de Fibroblastos/farmacología , Hipoglucemiantes/farmacología , Músculo Esquelético/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Factor 4 de Crecimiento de Fibroblastos/administración & dosificación , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/administración & dosificación , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/metabolismo , Inflamación , Resistencia a la Insulina , Ligandos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Músculo Esquelético/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Comunicación Paracrina , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos
13.
Biotechnol J ; 15(2): e1900203, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31556248

RESUMEN

Polyethylene glycol modification (PEGylation) can enhance the pharmacokinetic properties of therapeutic proteins by the attachment of polyethylene glycol (PEG) to the surface of a protein to shield the protein surface from proteolytic degradation and limit aggregation. However, current PEGylation strategies often reduce biological activity, potentially as a result of steric hindrance of PEG. Overall, there are no structure-based guidelines for selection of conjugate sites that retain optimal biological activity with improved pharmacokinetic properties. In this study, site-specific PEGylation based on the FGF2-FGFR1-heparin complex structure is performed. The effects of the conjugate sites on protein function are investigated by measuring the receptor/heparin binding affinities of the modified proteins and performing assays to measure cell-based bio-activity and in vivo stability. Comprehensive analysis of these data demonstrates that PEGylation of FGF2 that avoids the binding sites for fibroblast growth factor receptor 1 (FGFR1) and heparin provides optimal pharmacokinetic enhancement with minimal losses to biological activity. Animal experiments demonstrate that PEGylated FGF2 exhibits greater efficacy in protecting against traumatic brain injury-induced brain damage and neurological functions than the non-modified FGF2. This rational structure-based PEGylation strategy for protein modification is expected to have a major impact in the area of protein-based therapeutics.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Heparina/metabolismo , Polietilenglicoles/química , Procesamiento Proteico-Postraduccional , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Sitios de Unión , Escherichia coli/genética , Escherichia coli/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/farmacocinética , Heparina/farmacocinética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Unión Proteica , Conformación Proteica , Proteolisis , Ratas , Ratas Sprague-Dawley , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Resonancia por Plasmón de Superficie
14.
Front Pharmacol ; 10: 1478, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31866871

RESUMEN

Insulin resistance is associated with a greatly increased risk of type 2 diabetes. Administration of fibroblast growth factor-1 (FGF-1) resulted in a marked improvement in insulin sensitivity. However, the underlying molecular mechanism whereby FGF-1 represses insulin resistance remains largely unknown. Here, we sought to delineate the role of FGF-1 in insulin resistance with respect to its anti-inflammatory capability. In this study, we found that FGF-1 had positive effects on glucose intolerance, hepatic lipid accumulation, and insulin resistance, while it markedly repressed cytokine secretion (TNF-α and IL-6) in serum and reduced liver inflammation in diet-induced obesity (DIO) mice. Further, FGF-1 treatment significantly represses TNF-α-induced insulin resistance in vitro and in vivo. These results indicate that FGF-1 likely ameliorates insulin resistance via a mechanism that is independent of its glucose-lowering activity. Subsequent experiments demonstrated that FGF-1 ameliorated insulin resistance, and inflammation was accompanied by decreased c-Jun N-terminal kinase (JNK) signaling. In addition, it is likely that FGF-1 impedes JNK phosphorylation via blocking the transforming growth factor-ß activated kinase 1 (TAK1) and TAK1 binding protein 1 (TAB1) interaction. These findings reveal that FGF-1 regulates insulin sensitivity and may represent an attractive therapeutic target for preventing the development of insulin resistance.

15.
Front Pharmacol ; 10: 1515, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31920680

RESUMEN

Endocrine fibroblast growth factor (FGF) 19 has been shown to be capable of maintaining bile acid (BA) homeostasis and thus hold promise to be a potential therapeutic agent for cholestasis liver disease. However, whether paracrine FGFs possess this BA regulatory activity remains to be determined. In our study, we identified that paracrine fibroblast growth factor 1 (FGF1) was selectively downregulated in the liver of alpha naphthylisothiocyanate (ANIT)-induced intrahepatic cholestasis mice, suggesting a pathological relevance of this paracrine FGF with abnormal BA metabolism. Therefore, we evaluated the effects of engineered FGF1 mutant - FGF1ΔHBS on the metabolism of hepatic BA and found that this protein showed a more potent inhibitory effect of BA biosynthesis than FGF19 without any hepatic mitogenic activity. Moreover, the chronic administration of FGF1ΔHBS protected liver against ANIT-induced injury by reducing hepatic BA accumulation. Taken together, these data suggest that FGF1ΔHBS may function as a potent therapeutic agent for intrahepatic cholestasis liver disease.

16.
EBioMedicine ; 48: 462-477, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31631034

RESUMEN

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.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Enfermedades Metabólicas/metabolismo , Comunicación Paracrina , Adipocitos/metabolismo , Animales , Biomarcadores , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/química , Factores de Crecimiento de Fibroblastos/genética , Expresión Génica , Humanos , Insulina/metabolismo , Masculino , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/etiología , Ratones , Modelos Moleculares , Comunicación Paracrina/efectos de los fármacos , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes , Relación Estructura-Actividad
17.
Drug Deliv ; 25(1): 1560-1569, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30043675

RESUMEN

Fibroblast growth factor-20 (FGF20) is a paracrine member of the FGF family that is preferentially expressed in the substantia nigra pars compacta (SNpc). Previous studies have demonstrated that FGF20 enhances the survival of dopaminergic neurons suggesting the potential use of FGF20 to treat Parkinson's disease (PD). However, the reduced solubility of the bacterial recombinant human FGF20 (rhFGF20) and the absence of efficient strategies to transport rhFGF20 across the blood-brain barrier (BBB) have halted its clinical application. In the present study, we have examined the efficiency of fuzing a small ubiquitin-related modifier (SUMO) to rhFGF20 to enhance its soluble expression and further investigated the efficacy of FUS-guided, rhFGF20-liposome transport across the BBB. We also examined the bioavailability and behavioral improvement in a 6-hydroxydopamine-lesioned rat model of PD following 2 weeks' FUS-liposomal combinatorial treatment. Our results showed that, in contrast with rhFGF20 or LIP-FGF20, the FUS-LIP-rhFGF20 treatment could significantly improve the apomorphine-induced rotations by protecting against the loss of dopaminergic neurons in the SNpc. Our Results suggest that our combinatorial method would help overcome key challenges that hinder the currently available methods for the use of rhFGF20 in PD treatment.


Asunto(s)
Factores de Crecimiento de Fibroblastos/administración & dosificación , Enfermedad de Parkinson/diagnóstico por imagen , Enfermedad de Parkinson/tratamiento farmacológico , Proteolípidos/administración & dosificación , Animales , Línea Celular Tumoral , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Liposomas , Masculino , Ratones , Células 3T3 NIH , Oxidopamina/toxicidad , Células PC12 , Enfermedad de Parkinson/metabolismo , Proteolípidos/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento , Ultrasonografía/métodos
18.
Cell Rep ; 20(7): 1717-1728, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28813681

RESUMEN

The recent discovery of metabolic roles for fibroblast growth factor 1 (FGF1) in glucose homeostasis has expanded the functions of this classically known mitogen. To dissect the molecular basis for this functional pleiotropy, we engineered an FGF1 partial agonist carrying triple mutations (FGF1ΔHBS) that diminished its ability to induce heparan sulfate (HS)-assisted FGF receptor (FGFR) dimerization and activation. FGF1ΔHBS exhibited a severely reduced proliferative potential, while preserving the full metabolic activity of wild-type FGF1 in vitro and in vivo. Hence, suboptimal FGFR activation by a weak FGF1-FGFR dimer is sufficient to evoke a metabolic response, whereas full FGFR activation by stable and sustained dimerization is required to elicit a mitogenic response. In addition to providing a physical basis for the diverse activities of FGF1, our findings will impact ongoing drug discoveries targeting FGF1 and related FGFs for the treatment of a variety of human diseases.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/química , Hepatocitos/efectos de los fármacos , Mitógenos/química , Receptores de Factores de Crecimiento de Fibroblastos/química , Células 3T3-L1 , Animales , Sitios de Unión , Línea Celular Tumoral , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Factor 1 de Crecimiento de Fibroblastos/genética , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Expresión Génica , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Mitógenos/genética , Mitógenos/metabolismo , Mitógenos/farmacología , Modelos Moleculares , Células 3T3 NIH , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Estabilidad Proteica , Ratas , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Mol Neurobiol ; 51(1): 32-42, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24930088

RESUMEN

Fibroblast growth factor 2 (FGF-2) has a neurotrophic effect on dopaminergic neurons in vitro and in vivo, and exhibits beneficial effects in animal models of neurodegenerative disorders such as Parkinson's disease (PD). The poor stability and short half-life of FGF-2, however, have hampered its clinical use for neurological diseases. In the present study, we modified native recombinant human FGF-2 (rhFGF-2) by covalently attaching polyethylene glycol (PEG) polymers, named PEGylation, to enhance its neuroprotection efficacy in 6-hydroxydopamine (6-OHDA)-induced model of PD. In vitro, PEG-rhFGF-2 performed better biostability in 6-OHDA-induced PC-12 cells than native rhFGF-2. The in vivo data showed that, compared with native rhFGF-2, PEGylated rhFGF-2 was more efficacious in preventing 6-OHDA-induced lesion upon tyrosine hydroxylase-positive neurons in the substantia nigra (SN), improving the apomorphine-induced rotational behavior and the 6-OHDA-induced decline in tissue concentration of dopamine (DA) and its metabolites. Importantly, our data showed that the superior pharmacological activity of PEGylated rhFGF-2 is probably due to its greater permeability through the blood-brain barrier and better in vivo stability compared to native rhFGF-2. The enhanced stability and bioavailability of PEGylated rhFGF-2 make this molecule a great therapeutic candidate for neurodegenerative diseases such as PD and mood disorders.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Factor 2 de Crecimiento de Fibroblastos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/fisiopatología , Polietilenglicoles/uso terapéutico , Proteínas Recombinantes/uso terapéutico , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Apomorfina/farmacología , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Conducta Animal/efectos de los fármacos , Disponibilidad Biológica , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Factor 2 de Crecimiento de Fibroblastos/farmacocinética , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Neostriado/patología , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/farmacología , Oxidopamina , Células PC12 , Enfermedad de Parkinson/patología , Polietilenglicoles/farmacocinética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacocinética , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología , Tirosina 3-Monooxigenasa/metabolismo
20.
J Chromatogr A ; 1327: 66-72, 2014 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-24411087

RESUMEN

PEGylation is a widely applied approach to improve the pharmacokinetic and pharmacodynamic properties of protein therapeutics. The current solution-phase PEGylation protocols often suffer from poor yield of homogeneously PEGylated bioactive products and hence fall short of being commercially attractive. To improve upon these techniques, here we developed a novel, solid-phase PEGylation methodology using a hydrophobic interaction chromatography (HIC) resin. Two variations of the HIC-based PEGylation are described that are tailored towards conjugation of proteins with hydrophobicity index above (lysozyme) and below (fibroblast growth factor 1, FGF-1) that of the mPEG-butyraldehyde (mPEG) chain used. In the case of lysozyme, the protein was first immobilized on the HIC, and the HIC-bound protein was then conjugated by passing over the column. In the case of FGF-1, the mPEG solution was first immobilized on the HIC, and the FGF-1 solution was then passed through the column. Circular dichroism (CD) spectroscopy demonstrated HIC-based PEGylation almost retained the secondary structures of proteins. Bioactivity assay showed that the recovery of activity of HIC-based PEGylated rhFGF-1 (i.e. 92%) was higher than that of liquid-phase PEGylated rhFGF-1 (i.e. 61%), while HIC-based PEGylated lysozyme showed the same activity recovery (i.e. 7%) as the liquid-phase PEGylated form. For specific proteins, the HIC-based solid-phase PEGylation maybe offer a more promising alternative than the current PEGylation methods and is expected to have a major impact in the area of protein-based therapeutics.


Asunto(s)
Polietilenglicoles/química , Cromatografía/métodos , Dicroismo Circular , Factor 1 de Crecimiento de Fibroblastos/análisis , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Muramidasa/análisis , Estructura Secundaria de Proteína , Proteínas Recombinantes/análisis , Soluciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA