Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.397
Filter
1.
Int J Mol Med ; 53(6)2024 06.
Article in English | MEDLINE | ID: mdl-38666537

ABSTRACT

Fibroblast growth factor (FGF)21 is a peptide hormone that improves mitochondrial function and energy metabolism, and the deficiency of its co­receptor ß­klotho (KLB) causes decreased FGF21 sensitivity. The present study examined whether the cardiac delivery of plasmids containing the KLB gene via ultrasound­targeted microbubble destruction (UTMD) enhances the efficacy of FGF21 against heart failure post­acute myocardial infarction (AMI). For this purpose, the levels of FGF21 in patients and rats with heart dysfunction post­infarction were determined using ELISA. Sprague­Dawley rats received the 3X UTMD­mediated delivery of KLB@cationic microbubbles (KLB@CMBs) 1 week following the induction of AMI. Echocardiography, histopathology and biochemical analysis were performed at 4 weeks following the induction of AMI. The results revealed that patients with heart failure post­infarction had higher serum FGF21 levels than the healthy controls. However, the downstream signal, KLB, but not α­klotho, was reduced in the heart tissues of rats with AMI. As was expected, treatment with FGF21 did not substantially attenuate heart remodeling post­infarction. It was found that decreased receptors KLB in the heart may result in the insensitivity to FGF21 treatment. In vivo, the UTMD technology­mediated delivery of KLB@CMBs to the heart significantly enhanced the effects of FGF21 administration on cardiac remodeling and mitochondrial dysfunction in the rats following infarction. The delivery of KLB to the heart by UTMD and the administration of FGF21 attenuated mitochondrial impairment and oxidative stress by activating nuclear factor erythroid 2­related factor 2 signals. On the whole, the present study demonstrates that the cardiac delivery of KLB significantly optimizes the cardioprotective effects of FGF21 therapy on adverse heart remodeling. UTMD appears a promising interdisciplinary approach with which to improve heart failure post­myocardial infarction.


Subject(s)
Fibroblast Growth Factors , Klotho Proteins , Microbubbles , Myocardial Infarction , Rats, Sprague-Dawley , Ventricular Remodeling , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Humans , Male , Rats , Ventricular Remodeling/drug effects , Female , Ultrasonic Waves , Myocardium/metabolism , Myocardium/pathology , Heart Failure/metabolism , Heart Failure/therapy
2.
Exp Clin Endocrinol Diabetes ; 132(3): 152-161, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38513652

ABSTRACT

BACKGROUND: Type 2 diabetes mellitus (T2DM) is a commonly observed complication associated with obesity. The effect of fibroblast growth factor 19 (FGF19), a promising therapeutic agent for metabolic disorders, on pancreatic ß cells in obesity-associated T2DM remains poorly understood. METHODS: Human pancreatic ß cells were cultured with high glucose (HG) and palmitic acid (PA), followed by treatment with FGF19. The cell proliferation, apoptosis, and insulin secretion were evaluated by CCK-8, qRT-PCR, ELISA, flow cytometry, and western blotting. The expression of the insulin receptor substrate (IRS)/glucose transporter (GLUT) pathway was evaluated. The interaction between FGF19 and IRS1 was predicted using the STRING database and verified by co-immunoprecipitation and immunofluorescence. The regulatory effects of the IRS1/GLUT4 pathway on human pancreatic ß cells were assessed by overexpressing IRS1 and silencing IRS1 and GLUT4. RESULTS: HG+PA treatment reduced the human pancreatic ß cell proliferation and insulin secretion and promoted cell apoptosis. However, FGF19 treatment restored these alterations and significantly increased the expressions of IRS1, GLUT1, and GLUT4 in the IRS/GLUT pathway. Furthermore, FGF19 and IRS1 were found to interact. IRS1 overexpression partially promoted the proliferation of pancreatic ß cells and insulin secretion through GLUT4. Additionally, the silencing of IRS1 or GLUT4 attenuated the therapeutic effects of FGF19. CONCLUSION: In conclusion, FGF19 partly promoted the proliferation and insulin secretion of human pancreatic ß cells and inhibited apoptosis by upregulating the IRS1/GLUT4 pathway. These findings establish a theoretical framework for the clinical utilization of FGF19 in the treatment of obesity-associated T2DM.


Subject(s)
Diabetes Mellitus, Type 2 , Fibroblast Growth Factors , Glucose Transporter Type 1 , Insulin Receptor Substrate Proteins , Insulin Secretion , Insulin-Secreting Cells , Obesity , Humans , Cell Proliferation/drug effects , Diabetes Mellitus, Type 2/complications , Fibroblast Growth Factors/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/physiology , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Obesity/etiology , Obesity/therapy , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , Glucose Transporter Type 1/metabolism , Cell Line, Tumor , Glucose/metabolism , Glucose/pharmacology
3.
Breast Cancer Res ; 26(1): 54, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553760

ABSTRACT

Fibroblast growth factors (FGFs) control various cellular functions through fibroblast growth factor receptor (FGFR) activation, including proliferation, differentiation, migration, and survival. FGFR amplification in ER + breast cancer patients correlate with poor prognosis, and FGFR inhibitors are currently being tested in clinical trials. By comparing three-dimensional spheroid growth of ER + breast cancer cells with and without FGFR1 amplification, our research discovered that FGF2 treatment can paradoxically decrease proliferation in cells with FGFR1 amplification or overexpression. In contrast, FGF2 treatment in cells without FGFR1 amplification promotes classical FGFR proliferative signaling through the MAPK cascade. The growth inhibitory effect of FGF2 in FGFR1 amplified cells aligned with an increase in p21, a cell cycle inhibitor that hinders the G1 to S phase transition in the cell cycle. Additionally, FGF2 addition in FGFR1 amplified cells activated JAK-STAT signaling and promoted a stem cell-like state. FGF2-induced paradoxical effects were reversed by inhibiting p21 or the JAK-STAT pathway and with pan-FGFR inhibitors. Analysis of patient ER + breast tumor transcriptomes from the TCGA and METABRIC datasets demonstrated a strong positive association between expression of FGF2 and stemness signatures, which was further enhanced in tumors with high FGFR1 expression. Overall, our findings reveal a divergence in FGFR signaling, transitioning from a proliferative to stemness state driven by activation of JAK-STAT signaling and modulation of p21 levels. Activation of these divergent signaling pathways in FGFR amplified cancer cells and paradoxical growth effects highlight a challenge in the use of FGFR inhibitors in cancer treatment.


Subject(s)
Breast Neoplasms , Signal Transduction , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/therapeutic use , Janus Kinases/metabolism , Janus Kinases/pharmacology , Janus Kinases/therapeutic use , STAT Transcription Factors/metabolism , STAT Transcription Factors/pharmacology , STAT Transcription Factors/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1 , Cell Proliferation , Fibroblast Growth Factors/pharmacology , Cell Line, Tumor
4.
Dev Growth Differ ; 66(3): 182-193, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38342985

ABSTRACT

Newts can regenerate functional elbow joints after amputation at the joint level. Previous studies have suggested the potential contribution of cells from residual tendon tissues to joint cartilage regeneration. A serum-free tissue culture system for tendons was established to explore cell dynamics during joint regeneration. Culturing isolated tendons in this system, stimulated by regeneration-related factors, such as fibroblast growth factor (FGF) and platelet-derived growth factor, led to robust cell migration and proliferation. Moreover, cells proliferating in an FGF-rich environment differentiated into Sox9-positive chondrocytes upon BMP7 introduction. These findings suggest that FGF-stimulated cells from tendons may aid in joint cartilage regeneration during functional elbow joint regeneration in newts.


Subject(s)
Bone Morphogenetic Protein 7 , Chondrocytes , Fibroblast Growth Factors , Animals , Cell Differentiation , Chondrocytes/metabolism , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/metabolism , Salamandridae/metabolism , Tendons/metabolism , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/pharmacology
5.
Am J Physiol Renal Physiol ; 326(4): F584-F599, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38299214

ABSTRACT

Cardiovascular disease (CVD) is the major cause of death in chronic kidney disease (CKD) and is associated with high circulating fibroblast growth factor (FGF)23 levels. It is unresolved whether high circulating FGF23 is a mere biomarker or pathogenically contributes to cardiomyopathy. It is also unknown whether the C-terminal FGF23 peptide (cFGF23), a natural FGF23 antagonist proteolyzed from intact FGF23 (iFGF23), retards CKD progression and improves cardiomyopathy. We addressed these questions in three murine models with high endogenous FGF23 and cardiomyopathy. First, we examined wild-type (WT) mice with CKD induced by unilateral ischemia-reperfusion and contralateral nephrectomy followed by a high-phosphate diet. These mice were continuously treated with intraperitoneal implanted osmotic minipumps containing either iFGF23 protein to further escalate FGF23 bioactivity, cFGF23 peptide to block FGF23 signaling, vehicle, or scrambled peptide as negative controls. Exogenous iFGF23 protein given to CKD mice exacerbated pathological cardiac remodeling and CKD progression, whereas cFGF23 treatment improved heart and kidney function, attenuated fibrosis, and increased circulating soluble Klotho. WT mice without renal insult placed on a high-phosphate diet and homozygous Klotho hypomorphic mice, both of whom develop moderate CKD and clear cardiomyopathy, were treated with cFGF23 or vehicle. Mice treated with cFGF23 in both models had improved heart and kidney function and histopathology. Taken together, these data indicate high endogenous iFGF23 is not just a mere biomarker but pathogenically deleterious in CKD and cardiomyopathy. Furthermore, attenuation of FGF23 bioactivity by cFGF23 peptide is a promising therapeutic strategy to protect the kidney and heart from high FGF23 activity.NEW & NOTEWORTHY There is a strong correlation between cardiovascular morbidity and high circulating fibroblast growth factor 23 (FGF23) levels, but causality was never proven. We used a murine chronic kidney disease (CKD) model to show that intact FGF23 (iFGF23) is pathogenic and contributes to both CKD progression and cardiomyopathy. Blockade of FGF23 signaling with a natural proteolytic product of iFGF23, C-terminal FGF23, alleviated kidney and cardiac histology, and function in three separate murine models of high endogenous FGF23.


Subject(s)
Cardiomyopathies , Renal Insufficiency, Chronic , Animals , Mice , Fibroblast Growth Factor-23 , Disease Models, Animal , Renal Insufficiency, Chronic/metabolism , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/metabolism , Biomarkers , Phosphates , Cardiomyopathies/drug therapy , Cardiomyopathies/complications
6.
Int J Biol Macromol ; 261(Pt 1): 129797, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38290625

ABSTRACT

FGF21 plays an active role in the treatment of type 2 diabetes, obesity, nonalcoholic fatty liver disease (NAFLD), and nonalcoholic steatohepatitis (NASH). However, the short half-life and poor stability of wild-type FGF21 limit its clinical application. Previous studies found that PEGylation can significantly increase the stability of FGF21. However, the uneven distribution of PEGylation sites in FGF21 makes it difficult to purify PEG-FGF21, thereby affecting its yield, purity, and activity. To obtain long-acting FGF21 with controlled site-specific modification, we mutated lysine residues in FGF21, resulting in PEGylation only at the N-terminus of FGF21 (mFGF21). In addition, we modified mFGF21 molecules with different PEG molecules and selected the PEG-mFGF21 moiety with the highest activity. The yield of PEG-mFGF21 in this study reached 1 g/L (purity >99 %), and the purification process was simple and efficient with strong quality controllability. The half-life of PEG-mFGF21 in rats reached 40.5-67.4 h. Pharmacodynamic evaluation in mice with high-fat, high-cholesterol- and methionine and choline deficiency-induced NASH illustrated that PEG-mFGF21 exhibited long-term efficacy in improving liver steatosis and reducing liver cell damage, inflammation, and fibrosis. Taken together, PEG-mFGF21 could represent a potential therapeutic drug for the treatment of NASH.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Mice , Rats , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/therapeutic use , Fibroblast Growth Factors/pharmacology , Obesity/drug therapy , Liver
7.
Mol Pharmacol ; 105(3): 179-193, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38238100

ABSTRACT

The circadian clock is an endogenous biochemical timing system that coordinates the physiology and behavior of organisms to earth's ∼24-hour circadian day/night cycle. The central circadian clock synchronized by environmental cues hierarchically entrains peripheral clocks throughout the body. The circadian system modulates a wide variety of metabolic signaling pathways to maintain whole-body metabolic homeostasis in mammals under changing environmental conditions. Endocrine fibroblast growth factors (FGFs), namely FGF15/19, FGF21, and FGF23, play an important role in regulating systemic metabolism of bile acids, lipids, glucose, proteins, and minerals. Recent evidence indicates that endocrine FGFs function as nutrient sensors that mediate multifactorial interactions between peripheral clocks and energy homeostasis by regulating the expression of metabolic enzymes and hormones. Circadian disruption induced by environmental stressors or genetic ablation is associated with metabolic dysfunction and diurnal disturbances in FGF signaling pathways that contribute to the pathogenesis of metabolic diseases. Time-restricted feeding strengthens the circadian pattern of metabolic signals to improve metabolic health and prevent against metabolic diseases. Chronotherapy, the strategic timing of medication administration to maximize beneficial effects and minimize toxic effects, can provide novel insights into linking biologic rhythms to drug metabolism and toxicity within the therapeutical regimens of diseases. Here we review the circadian regulation of endocrine FGF signaling in whole-body metabolism and the potential effect of circadian dysfunction on the pathogenesis and development of metabolic diseases. We also discuss the potential of chrononutrition and chronotherapy for informing the development of timing interventions with endocrine FGFs to optimize whole-body metabolism in humans. SIGNIFICANCE STATEMENT: The circadian timing system governs physiological, metabolic, and behavioral functions in living organisms. The endocrine fibroblast growth factor (FGF) family (FGF15/19, FGF21, and FGF23) plays an important role in regulating energy and mineral metabolism. Endocrine FGFs function as nutrient sensors that mediate multifactorial interactions between circadian clocks and metabolic homeostasis. Chronic disruption of circadian rhythms increases the risk of metabolic diseases. Chronological interventions such as chrononutrition and chronotherapy provide insights into linking biological rhythms to disease prevention and treatment.


Subject(s)
Circadian Clocks , Metabolic Diseases , Humans , Animals , Circadian Rhythm/genetics , Circadian Clocks/genetics , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Metabolic Diseases/metabolism , Energy Metabolism , Mammals/metabolism
8.
Climacteric ; 27(3): 305-313, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38275172

ABSTRACT

OBJECTIVES: This study aimed to investigate the effect and the mechanism of recombinant human fibroblast growth factor 18 (rhFGF18) on postmenopausal osteoporosis. METHODS: The effect of rhFGF18 on the proliferation and apoptosis of osteoblasts and the mechanism underlying such an effect was evaluated using an oxidative stress model of the MC3T3-E1 cell line. Furthermore, ovariectomy was performed on ICR mice to imitate estrogen-deficiency postmenopausal osteoporosis. Bone metabolism and bone morphological parameters in the ovariectomized (OVX) mice were evaluated. RESULTS: The results obtained from the cell model showed that FGF18 promoted MC3T3-E1 cell proliferation by activating the extracellular signal-regulated kinase (ERK) and p38 instead of c-Jun N-terminal kinase (JNK). FGF18 also prevented cells from damage inflicted by oxidative stress via inhibition of apoptosis. After FGF18 administration, the expression level of anti-apoptotic protein Bcl-2 in the mice was upregulated, whereas those of the pro-apoptotic proteins Bax and caspase-3 were downregulated. Administering FGF18 also improved bone metabolism and bone morphological parameters in OVX mice. CONCLUSIONS: FGF18 could effectively prevent bone loss in OVX mice by enhancing osteoblastogenesis and protecting osteoblasts from oxidative stress-induced apoptosis.


Subject(s)
Apoptosis , Cell Proliferation , Disease Models, Animal , Fibroblast Growth Factors , Osteoblasts , Osteoporosis, Postmenopausal , Ovariectomy , Oxidative Stress , Recombinant Proteins , Animals , Fibroblast Growth Factors/pharmacology , Mice , Female , Apoptosis/drug effects , Recombinant Proteins/pharmacology , Osteoblasts/drug effects , Humans , Oxidative Stress/drug effects , Osteoporosis, Postmenopausal/prevention & control , Cell Proliferation/drug effects , Mice, Inbred ICR , Cell Line
9.
Gene Ther ; 31(3-4): 95-104, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37699965

ABSTRACT

Fibroblast growth factor 21 (FGF21) has been developed as a potential therapeutic agent for metabolic syndromes. Moreover, FGF21 is considered a pro-longevity hormone because transgenic mice overexpressing FGF21 display extended lifespan, raising the possibility of using FGF21 to promote healthy aging. We recently showed that visceral fat directed FGF21 gene therapy improves metabolic and immune health in insulin resistant BTBR mice. Here, we used a fat directed rAAV-FGF21 vector in 17-month-old female mice to investigate whether long-term FGF21 gene transfer could mitigate aging-related functional decline. Animals with FGF21 treatment displayed a steady, significant lower body weight over 7-month of the study compared to age-matched control mice. FGF21 treatment reduced adiposity and increased relative lean mass and energy expenditure associated with almost 100 folds higher serum level of FGF21. However, those changes were not translated into benefits on muscle function and did not affect metabolic function of liver. Overall, we have demonstrated that a single dose of fat-directed AAV-FGF21 treatment can provide a sustainable, high serum level of FGF21 over long period of time, and mostly influences adipose tissue homeostasis and energy expenditure. High levels of FGF21 alone in aged mice is not sufficient to improve liver or muscle functions.


Subject(s)
Adipose Tissue , Liver , Mice , Female , Animals , Adipose Tissue/metabolism , Liver/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Mice, Transgenic , Genetic Therapy
10.
Exp Brain Res ; 242(1): 25-32, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37910178

ABSTRACT

Parkinson's disease (PD) is one of the most common and complex Neurodegeneration, with an inherited metabolic disorder. Fibroblast growth factor 21 (FGF21), an endocrine hormone that belongs to the fibroblast growth factor superfamily, plays an extensive role in metabolic regulation. However, our understandings of the specific function and mechanisms of FGF21 on PD are still quite limited. Here, we aimed to elucidate the actions and the underlying mechanisms of FGF21 on dopaminergic neurodegeneration using cellular models of parkinsonism. To investigate the effects of FGF21 on dopaminergic neurodegeneration in vitro, proteasome impairment models of PD were utilized. Human dopaminergic neuroblastoma SH-SY5Y cells were treated with the proteasome inhibitor lactacystin (5 µmol/L) for 12 h, then with 50 ng/ml FGF-21 with or without 5 mmol/L of 3-methyladenine.The cells were dissected to assess alterations in autophagy using immunofluorescence, immunoblotting and electron microscopy assays. Our data indicate that FGF21 prevents dopaminergic neuron loss and shows beneficial effects against proteasome impairment induced PD syndrome, indicating it might be a potent candidate for developing novel drugs to deal with PD.


Subject(s)
Neuroblastoma , Parkinson Disease , Humans , Autophagy , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Fibroblast Growth Factors/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Proteasome Endopeptidase Complex/metabolism
11.
Hepatol Int ; 18(2): 610-622, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37864726

ABSTRACT

BACKGROUND & AIMS: Combination immunotherapy refers to the use of immune checkpoint inhibitors (ICI) and molecular-targeted agents (MTA), which have recently been approved for the treatment of advanced hepatocellular carcinoma (HCC). Owing to its relatively low antitumor effect (up to 30%), sequential therapy following ICIs treatment is required in patients with HCC. This study aimed to determine the impact of MTAs on the tumor immune microenvironment (TIME). METHODS: We established immune syngeneic orthotopic HCC mouse models using Hep-55.1C and Hep-53.4, and treated them with MTAs (lenvatinib, sorafenib, regorafenib, cabozantinib, and DC101 as anti-vascular endothelial growth factor receptor-2 antibodies, and AZD4547 as a fibroblast growth factor receptor (FGFR)-1/2/3/4 inhibitor) for 2 weeks. Subsequently, alterations in the TIME caused by MTAs were evaluated using immunohistochemistry (antibodies for CD3, CD8, Foxp3, Granzyme B, Arginase-1, NK1.1, F4/80, CD11c, PD-1, and PD-L1). We conducted RNA-seq analysis using lenvatinib- and AZD4547-treated tumors. To confirm the clinical relevance of these findings, we analyzed the transcriptome data of human HCC cells (MHCC-97H) treated with various concentrations of lenvatinib for 24 h using RNA-seq data from the Gene Expression Omnibus database. RESULTS: The number of Foxp3- and F4/80-positive cells in the TIME was decreased in many MTAs. Cabozantinib increased the numbers in NK1.1-, Granzyme B, and CD11c-positive cells. Lenvatinib and AZD4547 increased the number of CD8, Granzyme B, and PD-L1-positive cells. Gene ontology enrichment analysis revealed that lipid metabolism-related genes were downregulated by lenvatinib and AZD4547. In total, 161 genes downregulated by FGFR inhibition in rodent models overlapped with those downregulated by lenvatinib in human HCC cells. CONCLUSIONS: In this study, we showed that cabozantinib activated the innate immune system, and lenvatinib and AZD4547, which commonly inhibit FGFR signaling, altered TIME to a hot immune state by downregulating lipid metabolism-related genes. These findings support the therapeutic use of combination immunotherapies.


Subject(s)
Anilides , Antineoplastic Agents , Benzamides , Carcinoma, Hepatocellular , Liver Neoplasms , Phenylurea Compounds , Piperazines , Pyrazoles , Pyridines , Quinolines , Animals , Mice , Humans , Carcinoma, Hepatocellular/pathology , B7-H1 Antigen , Granzymes/pharmacology , Granzymes/therapeutic use , Liver Neoplasms/pathology , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Immunosuppressive Agents/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Forkhead Transcription Factors/pharmacology , Forkhead Transcription Factors/therapeutic use , Tumor Microenvironment
12.
Prostate ; 84(1): 100-110, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37796107

ABSTRACT

BACKGROUND: Androgen receptor (AR) pathway inhibition remains the cornerstone for prostate cancer therapies. However, castration-resistant prostate cancer (CRPC) tumors can resist AR signaling inhibitors through AR amplification and AR splice variants in AR-positive CRPC (ARPC), and conversion to AR-null phenotypes, such as double-negative prostate cancer (DNPC) and small cell or neuroendocrine prostate cancer (SCNPC). We have shown previously that DNPC can bypass AR-dependence through fibroblast growth factor receptor (FGFR) signaling. However, the role of the FGFR pathway in other CRPC phenotypes has not been elucidated. METHODS: RNA-Seq analysis was conducted on patient metastases, LuCaP patient-derived xenograft (PDX) models, and CRPC cell lines. Cell lines (C4-2B, VCaP, and 22Rv1) and ex vivo LuCaP PDX tumor cells were treated with enzalutamide (ENZA) and FGFR inhibitors (FGFRi) alone or in combination and sensitivity was determined using cell viability assays. In vivo efficacy of FGFRi in ARPC, DNPC, and SCNPC were evaluated using PDX models. RESULTS: RNA-Seq analysis of FGFR signaling in metastatic specimens, LuCaP PDX models, and CRPC cell lines revealed significant FGF pathway activation in AR-low PC (ARLPC), DNPC, and SCNPC tumors. In vitro/ex vivo analysis of erdafitinib and CH5183284 demonstrated robust and moderate growth suppression of ARPC, respectively. In vivo studies using four ARPC PDX models showed that combination ENZA and CH5183284 significantly suppressed tumor growth. Additional in vivo studies using four ARPC PDX models revealed that erdafitinib monotherapy was as effective as ENZA in suppressing tumor growth, and there was limited combination benefit. Furthermore, two of three DNPC models and two of four SCNPC models responded to CH5183284 monotherapy, suggesting FGFRi responses were model dependent. RNA-Seq and gene set enrichment analysis of end-of-study ARPC tumors treated with FGFRi displayed decreased expression of E2F and MYC target genes and suppressed G2M checkpoint genes, whereas end-of-study SCNPC tumors had heterogeneous transcriptional responses. CONCLUSIONS: Although FGFRi treatments suppressed tumor growth across CRPC phenotypes, our analyses did not identify a single pathway or biomarker that would identify tumor response to FGFRi. This is very likely due to the array of FGFR1-4 expression and tumor phenotypes present in CRPC. Nevertheless, our data nominate the FGFR pathway as a clinically actionable target that promotes tumor growth in diverse phenotypes of treatment-refractory metastatic CRPC.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Male , Humans , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/pharmacology , Androgens/pharmacology , Signal Transduction , Cell Line, Tumor , Nitriles/pharmacology
13.
Physiol Behav ; 272: 114368, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37805134

ABSTRACT

Pharmacological administration of fibroblast growth factor 21 (FGF21) alters food choice, including that it decreases the consumption of sucrose and other sweet tastants. Conversely, endogenous secretion of FGF21 by the liver is modulated by diet, such that plasma FGF21 is increased after eating foods that have a low dietary protein: total energy (P: E) ratio. Together, these findings suggest a strategy to promote healthy eating, in which the macronutrient content of a pre-load diet could reduce the consumption of sweet desserts in sated mice. Here, we tested the prediction that individuals maintained on a low P: E diet, and offered a highly palatable sweet 'dessert' following a pre-load meal, would eat less of the sugary snack compared to controls-due to increased FGF21 signaling. In addition to decreasing sweet intake, FGF21 increases the consumption of dietary protein. Thus, we predicted that individuals maintained on the low P: E diet, and offered a very high-protein pellet as 'dessert' or snack after a meal, would eat more of the high protein pellet compared to controls, and that this depends on FGF21. We tested this in C57Bl/6J, and liver-specific FGF21-null (FGF21ΔL) null male and female mice and littermate controls. Contrary to expectation, eating a low protein pre-load did not reduce the later consumption of a sweet solution in either males or females, despite robustly increasing plasma FGF21. Rather, eating the low protein pre-load increased later consumption of a high protein pellet. This was more apparent among males and was abrogated in the FGF21ΔL mice. We conclude that physiologic induction of hepatic FGF21 by a low protein pre-load diet is not sufficient to reduce the consumption of sweet desserts, though it effectively increases the subsequent intake of dietary protein in male mice.


Subject(s)
Diet, Protein-Restricted , Fibroblast Growth Factors , Male , Female , Mice , Animals , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Liver/metabolism , Dietary Proteins/pharmacology
14.
Biomed Pharmacother ; 168: 115737, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37862975

ABSTRACT

AIMS/HYPOTHESIS: Fibroblast growth factor 21 (FGF21) is a hepatokine with pleiotropic effects on glucose and lipid metabolic homeostasis. Here, we aimed to elucidate the mechanisms underlying the protective effects of FGF21 on L-lactate homeostasis and liver lesions in a type 1 diabetes mellitus (T1DM) mice model. METHODS: Six-week-old male C57BL/6 mice were divided into control, T1DM, and FGF21 groups. We also examined hepatic apoptotic signaling and functional indices in wild-type and hydroxycarboxylic acid receptor 1 (HCA1) knockout mice with T1DM or long-term L-lactate exposure. After preincubation of high glucose- or L-lactate treated hepatic AML12 cells, L-lactate uptake, apoptosis, and monocarboxylic acid transporter 2 (MCT2) expression were investigated. RESULTS: In a mouse model of T1DM, hepatic FGF21 expression was downregulated by approximately 1.5-fold at 13 weeks after the hyperglycemic insult. In vivo administration of exogenous FGF21 (2 mg/kg) to diabetic or L-lactate-infused mice significantly prevented hepatic oxidative stress and apoptosis by activating extracellular signal-regulated kinase (ERK)1/2, p38 mitogen-activated protein kinase (MAPK) and AMP-activated protein kinase (AMPK) pathways. HCA1-KO mice were less susceptible to diabetes- and L-lactate-induced hepatic apoptosis and dysfunction. In addition, inhibition of PI3K-mTOR activity revealed that FGF21 prevented L-lactate-induced Cori cycle alterations and hepatic apoptosis by upregulating MCT2 protein translation. CONCLUSIONS/INTERPRETATION: These results demonstrate that L-lactate homeostasis may be a therapeutic target for T1DM-related hepatic dysfunction. The protective effects of FGF21 on hepatic damage were associated with its ability to ameliorate MCT2-dependent Cori cycle alterations and prevent HCA1-mediated inhibition of ERK1/2, p38 MAPK, and AMPK signaling.


Subject(s)
Diabetes Mellitus, Type 1 , Mice , Male , Animals , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , AMP-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , Liver , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/metabolism , Disease Models, Animal , Glucose/metabolism , Homeostasis , Apoptosis , Mice, Knockout
15.
J Biol Chem ; 299(9): 105127, 2023 09.
Article in English | MEDLINE | ID: mdl-37544647

ABSTRACT

Diabetic keratopathy, commonly associated with a hyperactive inflammatory response, is one of the most common eye complications of diabetes. The peptide hormone fibroblast growth factor-21 (FGF-21) has been demonstrated to have anti-inflammatory and antioxidant properties. However, whether administration of recombinant human (rh) FGF-21 can potentially regulate diabetic keratopathy is still unknown. Therefore, in this work, we investigated the role of rhFGF-21 in the modulation of corneal epithelial wound healing, the inflammation response, and oxidative stress using type 1 diabetic mice and high glucose-treated human corneal epithelial cells. Our experimental results indicated that the application of rhFGF-21 contributed to the enhancement of epithelial wound healing. This treatment also led to advancements in tear production and reduction in corneal edema. Moreover, there was a notable reduction in the levels of proinflammatory cytokines such as TNF-α, IL-6, IL-1ß, MCP-1, IFN-γ, MMP-2, and MMP-9 in both diabetic mouse corneal epithelium and human corneal epithelial cells treated with high glucose. Furthermore, we found rhFGF-21 treatment inhibited reactive oxygen species production and increased levels of anti-inflammatory molecules IL-10 and SOD-1, which suggests that FGF-21 has a protective role in diabetic corneal epithelial healing by increasing the antioxidant capacity and reducing the release of inflammatory mediators and matrix metalloproteinases. Therefore, we propose that administration of FGF-21 may represent a potential treatment for diabetic keratopathy.


Subject(s)
Corneal Diseases , Diabetes Complications , Diabetes Mellitus, Experimental , Epithelium, Corneal , Fibroblast Growth Factors , Inflammation Mediators , Oxidative Stress , Wound Healing , Animals , Humans , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Corneal Diseases/complications , Corneal Diseases/drug therapy , Corneal Diseases/metabolism , Diabetes Complications/drug therapy , Diabetes Complications/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Epithelium, Corneal/drug effects , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/therapeutic use , Glucose/adverse effects , Glucose/metabolism , Inflammation Mediators/metabolism , Matrix Metalloproteinases/metabolism , Oxidative Stress/drug effects , Wound Healing/drug effects
16.
Biol Open ; 12(8)2023 08 15.
Article in English | MEDLINE | ID: mdl-37530863

ABSTRACT

The mesodermal precursor populations for different internal organ systems are specified during gastrulation by the combined activity of extracellular signaling systems such as BMP, Wnt, Nodal and FGF. The BMP, Wnt and Nodal signaling requirements for the differentiation of specific mesoderm subtypes in mammals have been mapped in detail, but how FGF shapes mesodermal cell type diversity is not precisely known. It is also not clear how FGF signaling integrates with the activity of other signaling systems involved in mesoderm differentiation. Here, we address these questions by analyzing the effects of targeted signaling manipulations in differentiating stem cell populations at single-cell resolution. We identify opposing functions of BMP and FGF, and map FGF-dependent and -independent mesodermal lineages. Stimulation with exogenous FGF boosts the expression of endogenous Fgf genes while repressing Bmp ligand genes. This positive autoregulation of FGF signaling, coupled with the repression of BMP signaling, may contribute to the specification of reproducible and coherent cohorts of cells with the same identity via a community effect, both in the embryo and in synthetic embryo-like systems.


Subject(s)
Fibroblast Growth Factors , Gastrulation , Animals , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Cell Differentiation/genetics , Mesoderm , Embryonic Stem Cells/metabolism , Mammals/metabolism
17.
Neuropeptides ; 101: 102367, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37506425

ABSTRACT

Alzheimer's disease (AD) is a multifactorial pathology marked by amyloid beta (Aß) accumulation, tau hyperphosphorylation, and progressive cognitive decline. Previous studies show that fibroblast growth factor 18 (FGF18) exerts a neuroprotective effect in experimental models of neurodegeneration; however, how it affects AD pathology remains unknown. This study aimed to ascertain the impact of FGF18 on the behavioral and neuropathological changes in the rat model of sporadic AD induced by intracerebroventricular (ICV) injection of streptozotocin (STZ). The rats were treated with FGF18 (0.94 and 1.88 pmol, ICV) on the 15th day after STZ injection. Their cognitive function was assessed in the Morris water maze and passive avoidance tests for 5 days from the 16th to the 21st days. Aß levels and histological signs of neurotoxicity were detected using the enzyme-linked immunosorbent assay (ELISA) assay and histopathological analysis of the brain, respectively. FGF18 mildly ameliorated the STZ-induced cognitive impairment; the Aß accumulation was reduced; and the neuronal damage including pyknosis and apoptosis was alleviated in the rat brain. This study highlights the promising therapeutic potential for FGF18 in managing AD.


Subject(s)
Alzheimer Disease , Rats , Animals , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/therapeutic use , Brain/metabolism , Memory Disorders/drug therapy , Disease Models, Animal , Streptozocin , Maze Learning
18.
Mol Cell Proteomics ; 22(8): 100594, 2023 08.
Article in English | MEDLINE | ID: mdl-37328066

ABSTRACT

Fibroblast growth factors (FGFs) are paracrine or endocrine signaling proteins that, activated by their ligands, elicit a wide range of health and disease-related processes, such as cell proliferation and the epithelial-to-mesenchymal transition. The detailed molecular pathway dynamics that coordinate these responses have remained to be determined. To elucidate these, we stimulated MCF-7 breast cancer cells with either FGF2, FGF3, FGF4, FGF10, or FGF19. Following activation of the receptor, we quantified the kinase activity dynamics of 44 kinases using a targeted mass spectrometry assay. Our system-wide kinase activity data, supplemented with (phospho)proteomics data, reveal ligand-dependent distinct pathway dynamics, elucidate the involvement of not earlier reported kinases such as MARK, and revise some of the pathway effects on biological outcomes. In addition, logic-based dynamic modeling of the kinome dynamics further verifies the biological goodness-of-fit of the predicted models and reveals BRAF-driven activation upon FGF2 treatment and ARAF-driven activation upon FGF4 treatment.


Subject(s)
Fibroblast Growth Factor 2 , Fibroblast Growth Factors , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Phosphorylation , Cell Proliferation , Mass Spectrometry
19.
Cells ; 12(8)2023 04 13.
Article in English | MEDLINE | ID: mdl-37190055

ABSTRACT

Diabetes mellitus contributes to 15-25% of all chronic foot ulcers. Peripheral vascular disease is a cause of ischemic ulcers and exacerbates diabetic foot disease. Cell-based therapies are viable options to restore damaged vessels and induce the formation of new vessels. Adipose-derived stem cells (ADSCs) have the potential for angiogenesis and regeneration because of their greater paracrine effect. Preclinical studies are currently using other forced enhancement techniques (e.g., genetic modification or biomaterials) to increase the efficacy of human ADSC (hADSC) autotransplantation. Unlike genetic modifications and biomaterials, many growth factors have been approved by the equivalent regulatory authorities. This study confirmed the effect of enhanced human ADSC (ehADSC)s with a cocktail of FGF and other pharmacological agents to promote wound healing in diabetic foot disease. In vitro, ehADSCs exhibited a long and slender spindle-shaped morphology and showed significantly increased proliferation. In addition, it was shown that ehADSCs have more functionalities in oxidative stress toleration, stem cell stemness, and mobility. In vivo, the local transplantation of 1.2 × 106 hADSCs or ehADSCs was performed in animals with diabetes induced by STZ. The ehADSC group showed a statistically decreased wound size and increased blood flow compared with the hADSC group and the sham group. Human Nucleus Antigen (HNA) positive cells were observed in some ADSC-transplanted animals. The ehADSC group showed a relatively higher portion of HNA-positive animals than the hADSC group. The blood glucose levels showed no significant difference among the groups. In conclusion, the ehADSCs showed a better performance in vitro, compared with conventional hADSCs. Additionally, a topical injection of ehADSCs into diabetic wounds enhanced wound healing and blood flow, while improving histological markers suggesting revascularization.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Foot , Humans , Rats , Animals , Streptozocin , Adipose Tissue , Diabetic Foot/therapy , Diabetic Foot/pathology , Fibroblast Growth Factors/pharmacology , Wound Healing/physiology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Experimental/pathology , Stem Cells , Biocompatible Materials/pharmacology
20.
Cell Metab ; 35(6): 1022-1037.e6, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37167965

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Mice , Animals , Male , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Fibroblast Growth Factor 4/therapeutic use , Rodentia/metabolism , Glucose/metabolism , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/therapeutic use , Fibroblast Growth Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...