Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.655
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1406442, 2024.
Article in English | MEDLINE | ID: mdl-39040677

ABSTRACT

Background: Diabetes ranks among the most widespread diseases globally, with the kidneys being particularly susceptible to its vascular complications. The identification of proteins for pathogenesis and novel drug targets remains imperative. This study aims to investigate roles of circulating inflammatory proteins in diabetic renal complications. Methods: Data on the proteins were derived from a genome-wide protein quantitative trait locus (pQTL) study, while data on diabetic renal complications came from the FinnGen study. In this study, proteome-wide Mendelian randomization (MR) and colocalization analyses were used to assess the relationship between circulating inflammatory proteins and diabetic renal complications. Results: MR approach indicated that elevated levels of interleukin 12B (IL-12B) (OR 1.691, 95%CI 1.179-2.427, P=4.34×10-3) and LIF interleukin 6 family cytokine (LIF) (OR 1.349, 95%CI 1.010-1.801, P=4.23×10-2) increased the risk of type 1 diabetes (T1D) with renal complications, while higher levels of fibroblast growth factor 19 (FGF19) (OR 1.202, 95%CI 1.009-1.432, P=3.93×10-2), fibroblast growth factor 23 (FGF23) (OR 1.379, 95%CI 1.035-1.837, P=2.82×10-2), C-C motif chemokine ligand 7 (CCL7) (OR 1.385, 95%CI 1.111-1.725, P=3.76×10-3), and TNF superfamily member 14 (TNFSF14) (OR 1.244, 95%CI 1.066-1.451, P=5.63×10-3) indicated potential risk factors for type 2 diabetes (T2D) with renal complications. Colocalization analysis supported these findings, revealing that most identified proteins, except for DNER, likely share causal variants with diabetic renal complications. Conclusion: Our study established associations between specific circulating inflammatory proteins and the risk of diabetic renal complications, suggesting these proteins as targets for further investigation into the pathogenesis and potential therapeutic interventions for T1D and T2D with renal complications.


Subject(s)
Diabetic Nephropathies , Mendelian Randomization Analysis , Proteome , Humans , Diabetic Nephropathies/blood , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/etiology , Proteome/metabolism , Proteome/analysis , Male , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Female , Quantitative Trait Loci , Genome-Wide Association Study , Inflammation/blood , Inflammation/metabolism , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Biomarkers/blood , Middle Aged
2.
J Cell Mol Med ; 28(14): e18551, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39054573

ABSTRACT

Despite numerous investigations on the influence of fibroblast growth factor 23 (FGF23), α-Klotho and FGF receptor-1 (FGFR1) on osteoporosis (OP), there is no clear consensus. Mendelian randomization (MR) analysis was conducted on genome-wide association studies (GWASs)-based datasets to evaluate the causal relationship between FGF23, α-Klotho, FGFR1 and OP. The primary endpoint was the odds ratio (OR) of the inverse-variance weighted (IVW) approach. Furthermore, we stably transfected FGF23-mimic or siRNA-FGF23 into human bone marrow mesenchymal stem cells (hBMSCs) in culture and determined its cell proliferation and the effects on osteogenic differentiation. Using MR analysis, we demonstrated a strong correlation between serum FGF23 levels and Heel- and femoral neck-BMDs, with subsequent ORs of 0.919 (95% CI: 0.860-0.983, p = 0.014) and 0.751 (95% CI: 0.587-0.962; p = 0.023), respectively. The expression levels of FGF23 were significantly increased in femoral neck of patients with OP than in the control cohort (p < 0.0001). Based on our in vitro investigation, after overexpression of FGF23, compared to the control group, the BMSC's proliferation ability decreased, the expression level of key osteogenic differentiation genes (RUNX2, OCN and OSX) significantly reduced, mineralized nodules and ALP activity significantly decreased. After silencing FGF23, it showed a completely opposite trend. Augmented FGF23 levels are causally associated with increased risk of OP. Similarly, FGF23 overexpression strongly inhibits the osteogenic differentiation of hBMSCs, thereby potentially aggravating the pathological process of OP.


Subject(s)
Cell Differentiation , Cell Proliferation , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Genome-Wide Association Study , Mendelian Randomization Analysis , Mesenchymal Stem Cells , Osteogenesis , Osteoporosis , Humans , Fibroblast Growth Factor-23/metabolism , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Cell Proliferation/genetics , Cell Differentiation/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Klotho Proteins/metabolism , Female , Glucuronidase/genetics , Glucuronidase/metabolism , Bone Density/genetics , Male , Middle Aged , Femur Neck/metabolism , Femur Neck/pathology
3.
BMC Pediatr ; 24(1): 418, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951759

ABSTRACT

PURPOSE: Metabolic bone disease of prematurity (MBDP) remains a significant cause of morbidity in extremely premature newborns. In high-risk patients, suspected diagnosis and subsequent treatment modifications, with limitations in terms of sensitivity and specificity, rely on low phosphorus levels and/or high levels of alkaline phosphatase (ALP). We investigated the potential of fibroblast growth factor-23 (FGF23) as an early marker for MBDP when measured at 3-4 weeks of life in at-risk patients. METHODS: A single-center prospective observational non-interventional study including preterm newborns of both sexes, with a gestational age of less than 32 weeks and/or a birth weight of less than 1500 g. In the standard biochemical screening for MBDP performed between 3 and 4 weeks of life within a nutritional profile, the determination of FGF23 was included along with other clinical and metabolic studies. The study was conducted at Marqués de Valdecilla University Hospital in Santander, Spain, from April 2020 to March 2021. Participants provided informed consent. Biochemical analyses were conducted using various platforms, and follow-up evaluations were performed at the discretion of neonatologists. Patients at high risk for MBDP received modifications in treatment accordingly. The sample was descriptively analyzed, presenting measures of central tendency and dispersion for continuous variables, and absolute numbers/percentages for categorical ones. Tests used included t-tests, Mann‒Whitney U tests, chi-square tests, logistic regressions, Pearson correlation, and ROC curve analysis (IBM SPSS Statistics version 19). Significance level: P < 0.05. RESULTS: In the study involving 25 at-risk premature newborns, it was found that 20% (n = 5) were diagnosed with MBDP. Three of these patients (60%) were identified as high-risk based on standard biochemical evaluation at 3-4 weeks of age, while the other two patients (40%) were diagnosed in subsequent weeks. However, in all 5 patients, measurement of FGF23 levels would allow for early identification and optimization of treatment before other markers become altered. Low levels of FGF23 at 3-4 weeks, even with normal phosphorus and ALP levels, indicate the need for modifications in nutritional supplementation. CONCLUSIONS: MBDP remains a significant concern in extremely premature newborns. Current diagnostic methods rely on limited biochemical markers. Early detection of low FGF23 levels enables timely interventions, potentially averting demineralization.


Subject(s)
Biomarkers , Bone Diseases, Metabolic , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Humans , Infant, Newborn , Female , Fibroblast Growth Factors/blood , Biomarkers/blood , Prospective Studies , Male , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/diagnosis , Bone Diseases, Metabolic/etiology , Infant, Premature, Diseases/diagnosis , Infant, Premature, Diseases/blood , Infant, Premature
4.
Rom J Morphol Embryol ; 65(2): 159-172, 2024.
Article in English | MEDLINE | ID: mdl-39020530

ABSTRACT

Obesity poses a significant and escalating challenge in contemporary society, increasing the risk of developing various metabolic disorders such as dyslipidemia, cardiovascular diseases, non-alcoholic fatty liver disease (NAFLD), type 2 diabetes, and certain types of cancer. The current array of therapeutic interventions for obesity remains insufficient, prompting a pressing demand for novel and more effective treatments. In response, scientific attention has turned to the fibroblast growth factor 21 (FGF21) due to its remarkable and diverse impacts on lipid, carbohydrate, and energy metabolism. This comprehensive review aims to delve into the multifaceted aspects of FGF21, encompassing its discovery, synthesis, functional roles, and potential as a biomarker and therapeutic agent, with a specific focus on its implications for NAFLD.


Subject(s)
Fibroblast Growth Factors , Non-alcoholic Fatty Liver Disease , Obesity , Humans , Fibroblast Growth Factors/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Obesity/complications , Animals
5.
Int J Biol Sci ; 20(9): 3412-3425, 2024.
Article in English | MEDLINE | ID: mdl-38993571

ABSTRACT

Chronic kidney disease (CKD) is linked to greater prevalence and rapid progression of calcific aortic valve disease (CAVD) characterized by valvular leaflet fibrosis and calcification. Fibroblast growth factor 23 (FGF23) level is elevated, and anti-aging protein Klotho is reduced in CKD patients. However, the roles of FGF23 and Klotho in the mechanism of aortic valve fibrosis and calcification remain unclear. We hypothesized that FGF23 mediates CKD-induced CAVD by enhancing aortic valve interstitial cell (AVIC) fibrosis and calcification, while soluble Klotho inhibits FGF23 effect. Methods and Results: In an old mouse model of CKD, kidney damages were accompanied by aortic valve thickening and calcification. FGF23 levels in plasma and aortic valve were increased, while Klotho levels were decreased. Recombinant FGF23 elevated the inflammatory, fibrogenic, and osteogenic activities in AVICs. Neutralizing antibody or shRNA targeting FGF23 suppressed the pathobiological activities in AVICs from valves affected by CAVD. FGF23 exerts its effects on AVICs via FGF receptor (FGFR)/Yes-associated protein (YAP) signaling, and inhibition of FGFR/YAP reduced FGF23's potency in AVICs. Recombinant Klotho downregulated the pathobiological activities in AVICs exposed to FGF23. Incubation of FGF23 with Klotho formed complexes and decreased FGF23's potency. Further, treatment of CKD mice with recombinant Klotho attenuated aortic valve lesions. Conclusion: This study demonstrates that CKD induces FGF23 accumulation, Klotho insufficiency and aortic valve lesions in old mice. FGF23 upregulates the inflammatory, fibrogenic and osteogenic activities in AVICs via the FGFR/YAP signaling pathway. Soluble Klotho suppresses FGF23 effect through molecular interaction and is capable of mitigating CKD-induced CAVD.


Subject(s)
Aortic Valve , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Glucuronidase , Klotho Proteins , Renal Insufficiency, Chronic , Klotho Proteins/metabolism , Fibroblast Growth Factor-23/metabolism , Animals , Renal Insufficiency, Chronic/metabolism , Glucuronidase/metabolism , Fibroblast Growth Factors/metabolism , Mice , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Male , Signal Transduction , Mice, Inbred C57BL , Humans , Aortic Valve Stenosis/metabolism , Disease Models, Animal
6.
Int J Dev Biol ; 68(2): 65-78, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39016374

ABSTRACT

During embryonic development, the vertebrate embryonic epiblast is divided into two parts including neural and superficial ectoderm. The neural plate border (NPB) is a narrow transitional area which locates between these parts and contains multipotent progenitor cells. Despite its small size, the cellular heterogeneity in this region produces specific differentiated cells. Signaling pathways, transcription factors, and the expression/repression of certain genes are directly involved in these differentiation processes. Different factors such as the Wnt signaling cascade, fibroblast growth factor (FGF), bone morphogenetic protein (BMP) signaling, and Notch, which are involved in various stages of the growth, proliferation, and differentiation of embryonic cells, are also involved in the determination and differentiation of neural plate border stem cells. Therefore, it is essential to consider the interactions and temporospatial coordination related to cells, tissues, and adjacent structures. This review examines our present knowledge of the formation of the neural plate border and emphasizes the requirement for interaction between different signaling pathways, including the BMP and Wnt cascades, the expression of its special target genes and their regulations, and the precise tissue crosstalk which defines the neural crest fate in the ectoderm at the early human embryonic stages.


Subject(s)
Bone Morphogenetic Proteins , Cell Differentiation , Gene Expression Regulation, Developmental , Neural Crest , Neural Plate , Signal Transduction , Neural Plate/metabolism , Neural Plate/embryology , Humans , Animals , Bone Morphogenetic Proteins/metabolism , Neural Crest/metabolism , Neural Crest/embryology , Ectoderm/metabolism , Ectoderm/embryology , Ectoderm/cytology , Wnt Signaling Pathway/physiology , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Germ Layers/metabolism , Germ Layers/cytology , Wnt Proteins/metabolism , Wnt Proteins/genetics
7.
Pharmacol Res ; 206: 107291, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38969274

ABSTRACT

Fibroblast growth factors (FGFs) act as proangiogenic and mitogenic cytokines in several cancers, including multiple myeloma (MM). Indeed, corrupted FGF autocrine and paracrine secretion induces an aberrant activation of the FGF receptor (FGFR) signaling sustaining cancer cell spreading and resistance to pharmacological treatments. Thus, FGF traps may represent a promising anti-cancer strategy to hamper the ligand-dependent activation of the FGF/FGFR system. We previously identified NSC12 as the first orally available small molecule FGF trap able to inhibit the growth and progression of several FGF-dependent tumor models. NSC12 is a pregnenolone derivative carrying a 1,1-bis-trifluoromethyl-1,3-propanediol chain in position 17 of the steroid nucleus. Investigation of structure-activity relationships (SARs) provided more potent and specific NSC12 steroid derivatives and highlighted that the C17-side chain is pivotal for the FGF trap activity. Here, a scaffold hopping approach allowed to obtain two FGF trap compounds (22 and 57) devoid of the steroid nucleus and able to efficiently bind FGF2 and to inhibit FGFR activation in MM cells. Accordingly, these compounds exert a potent anti-tumor activity on MM cell lines both in vitro and in vivo and on MM patient-derived primary cells, strongly affecting the survival of both proteasome-inhibitor sensitive and resistant MM cells. These results propose a new therapeutic option for relapsed/refractory MM patients and set the bases for the development of novel FGF traps prone to chemical diversification to be used in the clinic for the treatment of those tumors in which the FGF/FGFR system plays a pivotal role, including MM.


Subject(s)
Antineoplastic Agents , Fibroblast Growth Factors , Multiple Myeloma , Receptors, Fibroblast Growth Factor , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Humans , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Cell Line, Tumor , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Structure-Activity Relationship , Drug Discovery , Mice , Fibroblast Growth Factor 2/metabolism
8.
Cell Stem Cell ; 31(7): 945-946, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38971145

ABSTRACT

Dattani et al.1 developed a method for inducing hypoblast-like cells from human naive pluripotent stem cells. They elucidated the requirement for FGF signaling in human hypoblast specialization at a specific time window, which was previously controversial.


Subject(s)
Fibroblast Growth Factors , Signal Transduction , Humans , Fibroblast Growth Factors/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Cell Differentiation
9.
PLoS One ; 19(7): e0299975, 2024.
Article in English | MEDLINE | ID: mdl-38959242

ABSTRACT

Skeletal muscle growth is an economically important trait in the cattle industry. Secreted muscle-derived proteins, referred to as myokines, have important roles in regulating the growth, metabolism, and health of skeletal muscle in human and biomedical research models. Accumulating evidence supports the importance of myokines in skeletal muscle and whole-body health, though little is known about the potential presence and functional significance of these proteins in cattle. This study evaluates and confirms that secreted proteins acidic and rich in cysteine (SPARC), fibroblast growth factor 21 (FGF-21), myostatin (MSTN), and decorin (DCN) are expressed and SPARC, FGF-21, and DCN are secreted by primary bovine satellite cells from 3- (BSC3; n = 3) and 11- (BSC11; n = 3) month -old commercial angus steers. Cells were cultured and collected at zero, 12, 24, and 48 hours to characterize temporal expression and secretion from undifferentiated and differentiated cells. The expression of SPARC was higher in the undifferentiated (p = 0.04) and differentiated (p = 0.07) BSC11 than BSC3. The same was observed with protein secretion from undifferentiated (p <0.0001) BSC11 compared to BSC3. Protein secretion of FGF-21 was higher in undifferentiated BSC11 (p < 0.0001) vs. BSC3. DCN expression was higher in differentiated BSC11 (p = 0.006) vs. BSC3. Comparing undifferentiated vs. differentiated BSC, MSTN expression was higher in differentiated BSC3 (p ≤ 0.001) for 0, 12, and 24 hours and in BSC11 (p ≤ 0.03) for 0, 12, 24, and 48 hours. There is also a change over time for SPARC expression (p ≤ 0.03) in undifferentiated and differentiated BSC and protein secretion (p < 0.0001) in undifferentiated BSC, as well as FGF-21 expression (p = 0.007) in differentiated BSC. This study confirms SPARC, FGF-21, and DCN are secreted, and SPARC, FGF-21, MSTN, and DCN are expressed in primary bovine muscle cells with age and temporal differences.


Subject(s)
Cell Differentiation , Decorin , Fibroblast Growth Factors , Osteonectin , Animals , Cattle , Osteonectin/metabolism , Osteonectin/genetics , Fibroblast Growth Factors/metabolism , Decorin/metabolism , Cells, Cultured , Male , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Aging/metabolism , Myostatin/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology
10.
Zhonghua Nan Ke Xue ; 30(1): 3-8, 2024 Jan.
Article in Chinese | MEDLINE | ID: mdl-39046407

ABSTRACT

The testis serves as the reproductive gland in male mammals, primarily tasked with the production of sperm and synthesis of androgens. A complex signaling network consisting of various cell types, including germ cells, Sertoli cells, and Leydig cells, supports the structure and maintains the function of the testis. Apart from the hypothalamic-pituitary-gonadal axis, various sex hormones and cytokines are also implicated in the regulation of testicular function. The fibroblast growth factor (FGF) represents a crucial class of active cytokines that stimulate cell proliferation, induce tissue differentiation, and govern organ development. This review summarizes the molecular mechanisms of FGF regulating testicular development and spermatogenesis and maintaining male fertility.


Subject(s)
Fibroblast Growth Factors , Spermatogenesis , Testis , Male , Spermatogenesis/physiology , Testis/metabolism , Testis/physiology , Humans , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/physiology , Animals , Reproduction/physiology
11.
Front Immunol ; 15: 1390453, 2024.
Article in English | MEDLINE | ID: mdl-38962005

ABSTRACT

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


Subject(s)
Fibroblast Growth Factors , Macrophages , Receptors, Fibroblast Growth Factor , Signal Transduction , Humans , Macrophages/immunology , Macrophages/metabolism , Fibroblast Growth Factors/metabolism , Animals , Receptors, Fibroblast Growth Factor/metabolism , Macrophage Activation/immunology , Inflammation/immunology , Inflammation/metabolism
12.
Cell Mol Life Sci ; 81(1): 309, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060446

ABSTRACT

The circadian clock system coordinates metabolic, physiological, and behavioral functions across a 24-h cycle, crucial for adapting to environmental changes. Disruptions in circadian rhythms contribute to major metabolic pathologies like obesity and Type 2 diabetes. Understanding the regulatory mechanisms governing circadian control is vital for identifying therapeutic targets. It is well characterized that chromatin remodeling and 3D structure at genome regulatory elements contributes to circadian transcriptional cycles; yet the impact of rhythmic chromatin topology in metabolic disease is largely unexplored. In this study, we explore how the spatial configuration of the genome adapts to diet, rewiring circadian transcription and contributing to dysfunctional metabolism. We describe daily fluctuations in chromatin contacts between distal regulatory elements of metabolic control genes in livers from lean and obese mice and identify specific lipid-responsive regions recruiting the clock molecular machinery. Interestingly, under high-fat feeding, a distinct interactome for the clock-controlled gene Dbp strategically promotes the expression of distal metabolic genes including Fgf21. Alongside, new chromatin loops between regulatory elements from genes involved in lipid metabolism control contribute to their transcriptional activation. These enhancers are responsive to lipids through CEBPß, counteracting the circadian repressor REVERBa. Our findings highlight the intricate coupling of circadian gene expression to a dynamic nuclear environment under high-fat feeding, supporting a temporally regulated program of gene expression and transcriptional adaptation to diet.


Subject(s)
Chromatin , Circadian Clocks , Fatty Acids , Liver , Mice, Inbred C57BL , Mice, Obese , Obesity , Animals , Chromatin/metabolism , Chromatin/genetics , Liver/metabolism , Mice , Circadian Clocks/genetics , Obesity/metabolism , Obesity/genetics , Fatty Acids/metabolism , Male , Diet, High-Fat/adverse effects , Chromatin Assembly and Disassembly , Circadian Rhythm/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Lipid Metabolism/genetics , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Gene Expression Regulation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
13.
Int J Mol Sci ; 25(14)2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39063026

ABSTRACT

Liraglutide, a glucagon-like peptide 1 analog used to treat type 2 diabetes and obesity, is a potential new treatment modality for bile acid (BA) diarrhea. Here, we show that administration of liraglutide significantly decreased total BAs, especially the primary BAs, including cholic acid, chenodeoxycholic acid, taurocholic acid, taurochenodeoxycholic acid, glycocholic acid, and ß-muricholic acid, in the liver and feces. In addition, liraglutide significantly decreased tryptophan metabolites, including L-tryptophan, serotonin, 5-hydroxy indole-3-acetic acid, L-kynurenine, and xanthurenic acid, in the colon, whereas it significantly increased indole-3-propionic acid. Moreover, the administration of liraglutide remarkably decreased the expression of apical sodium-dependent bile acid transporter, which mediates BA uptake across the apical brush border member in the ileum, ileal BA binding protein, and fibroblast growth factor 15 in association with decreased expression of the BA-activated nuclear receptor farnesoid X receptor and the heteromeric organic solute transporter Ostα/ß, which induces BA excretion, in the ileum. Liraglutide acutely decreased body weight and blood glucose levels in association with decreases in plasma insulin and serotonin levels in food-deprived mice. These findings suggest the potential of liraglutide as a novel inhibitor of primary BAs and serotonin in the colon.


Subject(s)
Bile Acids and Salts , Colon , Glucagon-Like Peptide-1 Receptor , Liraglutide , Serotonin , Animals , Liraglutide/pharmacology , Serotonin/metabolism , Bile Acids and Salts/metabolism , Mice , Colon/metabolism , Colon/drug effects , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Male , Organic Anion Transporters, Sodium-Dependent/metabolism , Fibroblast Growth Factors/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Tryptophan/metabolism , Tryptophan/pharmacology , Tryptophan/analogs & derivatives , Mice, Inbred C57BL , Ileum/metabolism , Ileum/drug effects , Liver/metabolism , Liver/drug effects , Cholic Acids , Membrane Transport Proteins , Symporters
14.
PeerJ ; 12: e17615, 2024.
Article in English | MEDLINE | ID: mdl-38948228

ABSTRACT

Background: Fibroblast growth factor 21 (FGF21) is a key hormone factor that regulates glucose and lipid homeostasis. Exercise may regulate its effects and affect disease states. Therefore, we sought to determine how exercise affects FGF21 concentrations in adults. Methods: The review was registered in the International Prospective Systematic Review (PROSPERO, CRD42023471163). The Cochrane Library, PubMed, and Web of Science databases were searched for studies through July 2023. Studies that assessed the effects of exercise training on FGF21 concentration in adults were included. The random effect model, data with standardized mean difference (SMD), and 95% confidence intervals (CI) were used to evaluate the pooled effect size of exercise training on FGF21. The risk of heterogeneity and bias were evaluated. A total of 12 studies involving 401 participants were included. Results: The total effect size was 0.3 (95% CI [-0.3-0.89], p = 0.33) when comparing participants who exercised to those who were sedentary. However, subgroup analysis indicated that concurrent exercise and a duration ≥10 weeks significantly decreased FGF21 concentrations with an effect size of -0.38 (95% CI [-0.74--0.01], p < 0.05) and -0.38 (95% CI [-0.63--0.13], p < 0.01), respectively. Conclusion: Concurrent exercise and longer duration may be more efficient way to decrease FGF21 concentrations in adults with metabolic disorder.


Subject(s)
Exercise , Fibroblast Growth Factors , Fibroblast Growth Factors/blood , Humans , Exercise/physiology , Adult
15.
Sci Rep ; 14(1): 16270, 2024 07 15.
Article in English | MEDLINE | ID: mdl-39009650

ABSTRACT

Steroid-induced osteonecrosis of the femoral head (SONFH) is the predominant cause of non-traumatic osteonecrosis of the femoral head (ONFH). Impaired blood supply and reduced osteogenic activity of the femoral head are the key pathogenic mechanisms of SONFH. Fibroblast growth factor 23 (FGF23) levels are not only a biomarker for early vascular lesions caused by abnormal mineral metabolism, but can also act directly on the peripheral vascular system, leading to vascular pathology. The aim of this study was to observe the role of FGF23 on bone microarchitecture and vascular endothelium, and to investigate activation of pyroptosis in SONFH. Lipopolysaccharide (LPS) combined with methylprednisolone (MPS) was applied for SONFH mouse models, and adenovirus was used to increase or decrease the level of FGF23. Micro-CT and histopathological staining were used to observe the structure of the femoral head, and immunohistochemical staining was used to observe the vascular density. The cells were further cultured in vitro and placed in a hypoxic environment for 12 h to simulate the microenvironment of vascular injury during SONFH. The effect of FGF23 on osteogenic differentiation was evaluated using alkaline phosphatase staining, alizarin red S staining and expression of bone formation-related proteins. Matrigel tube formation assay in vitro and immunofluorescence were used to detect the ability of FGF23 to affect endothelial cell angiogenesis. Steroids activated the pyroptosis signaling pathway, promoted the secretion of inflammatory factors in SONFH models, led to vascular endothelial dysfunction and damaged the femoral head structure. In addition, FGF23 inhibited the HUVECs angiogenesis and BMSCs osteogenic differentiation. FGF23 silencing attenuated steroid-induced osteonecrosis of the femoral head by inhibiting the pyroptosis signaling pathway, and promoting osteogenic differentiation of BMSCs and angiogenesis of HUVECs in vitro.


Subject(s)
Femur Head Necrosis , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Osteogenesis , Pyroptosis , Pyroptosis/drug effects , Fibroblast Growth Factor-23/metabolism , Animals , Femur Head Necrosis/chemically induced , Femur Head Necrosis/metabolism , Femur Head Necrosis/pathology , Mice , Fibroblast Growth Factors/metabolism , Osteogenesis/drug effects , Humans , Femur Head/pathology , Femur Head/metabolism , Disease Models, Animal , Methylprednisolone/pharmacology , Male , Lipopolysaccharides/toxicity , Human Umbilical Vein Endothelial Cells/metabolism , Cell Differentiation , Steroids/pharmacology
16.
Rev Assoc Med Bras (1992) ; 70(6): e20231496, 2024.
Article in English | MEDLINE | ID: mdl-39045952

ABSTRACT

OBJECTIVE: The objective of this study was to determine serum fibroblast growth factor-23 levels in preeclampsia, eclampsia, gestational hypertension, and the presence of fetal growth restriction subgroups. METHODS: A total of 55 pregnant women with planned cesarean section were included in this cross-sectional study. They were divided into two groups, namely, control (25) and gestational hypertensive disease (30). The gestational hypertensive disease group was evaluated by dividing it into three subgroups (preeclampsia, eclampsia, and gestational hypertension) according to the clinical and laboratory findings of the disease and two subgroups (presence of fetal growth restriction and absence of fetal growth restriction) according to the birth weight percentile. Demographic parameters, obstetric history, physical examination findings, and laboratory values were evaluated. RESULTS: Demographic parameters and obstetric history were similar between the two groups, while gestational week of delivery was lower in the gestational hypertensive disease group (p=0.002). Laboratory parameters and serum fibroblast growth factor-23 (pg/mL) values were similar between the two groups. In the subgroup analysis for gestational hypertension, preeclampsia, and eclampsia, there was no statistically significant difference in serum fibroblast growth factor-23 levels between gestational hypertension, preeclampsia, eclampsia, and control groups. In the subgroup analysis based on the presence of fetal growth restriction, serum fibroblast growth factor-23 levels were similar to the control group in the gestational hypertensive disease absence of fetal growth restriction, while serum fibroblast growth factor-23 levels and serum calcium levels were statistically significantly lower in the gestational hypertensive disease with the presence of fetal growth restriction (p=0.044 and p<0.001, respectively). CONCLUSION: Serum fibroblast growth factor-23 levels are similar between pregnancies complicated with gestational hypertensive disease and normotensive pregnancies. However, serum fibroblast growth factor-23 levels were found to be lower in pregnancies complicated with gestational hypertensive disease with the presence of fetal growth restriction.


Subject(s)
Biomarkers , Fetal Growth Retardation , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Hypertension, Pregnancy-Induced , Pre-Eclampsia , Humans , Female , Pregnancy , Fetal Growth Retardation/blood , Cross-Sectional Studies , Adult , Hypertension, Pregnancy-Induced/blood , Fibroblast Growth Factors/blood , Fibroblast Growth Factor-23/blood , Biomarkers/blood , Pre-Eclampsia/blood , Case-Control Studies , Young Adult , Gestational Age , Eclampsia/blood
17.
Eur J Med Genet ; 70: 104958, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950880

ABSTRACT

BACKGROUND AND OBJECTIVE: X-linked hypophosphatemic rickets (XLH) is due to loss-of-function mutations in the phosphate-regulating endopeptidase homologue on the X chromosome (PHEX) that lead to increased fibroblast growth factor 23 (FGF23) production. FGF23 excess causes renal phosphate wasting and insufficient 1,25-dihydroxyvitamin D (1,25(OH)2D) synthesis with reduced intestinal phosphate absorption, ultimately resulting in chronic hypophosphatemia. Children with XLH show typical skeletal lesions of rickets, deformities of the lower limbs, stunted growth with disproportionate short stature, bone pain, and physical dysfunctions. Burosumab, a fully human IgG1 monoclonal antibody that binds to FGF23 to inhibit its activity, is more effective to improve the biochemical and clinical signs of XLH than conventional treatment with phosphate supplements and vitamin D active metabolites. Data on adolescents with XLH during the transition period to young adulthood are few. In this prospective case series, we aimed to assess safety and efficacy of burosumab in adolescents with XLH who discontinued long-term conventional therapy. METHODS: Five Caucasian adolescents (4 males, 1 female; mean age 15.4 ± 1.5 years) with XLH were recruited and switched from conventional treatment to burosumab (0.8-1.2 mg/kg, s. c. QW2). Burosumab was continued for 12-48 months and, once discontinued, patients were followed-up for 6-12 months. In all patients, serum calcium, phosphate, alkaline phosphatase (ALP), parathyroid hormone (PTH), and 1,25(OH)2D levels, and renal tubular reabsorption of phosphate (TmP/GFR) values were assessed at entry and during burosumab. Intact FGF23 plasma levels were measured at entry. Patient-reported outcomes (PROs) were assessed at entry and every 3-6 months to evaluate the impact of low extremity pain, stiffness, and difficulties performing daily activities. RESULTS: At entry, all patients showed hypophosphatemia, increased intact FGF23 levels, reduced TmP/GFR, insufficient 1,25(OH)2D levels, and in four out of five increased ALP levels. Two patients had radiological signs of rickets. During burosumab, all patients showed a significant increase in serum phosphate and 1,25(OH)2D levels, and in TmP/GFR values (P < 0.05 - P < 0.0001). Serum ALP levels significantly declined (P < 0.05) to normal values. No changes of serum calcium and PTH levels (PNS) were found during burosumab. PROs significantly improved (P < 0.02 - P < 0.0001) in all patients. Four patients discontinued burosumab when they turned 18 or 19, whereas one continued the treatment since he was still younger than 18 during the study period. Four patients who suspended burosumab showed a rapid decline in serum phosphate and 1,25(OH)2D levels and in TmP/GFR values; serum ALP levels increased, and PROs progressively worsened with a significant reduction in quality of life. These consequences were not observed in the patient who continued burosumab treatment. DISCUSSION: Our data showed that conventional treatment improved only in part the signs and symptoms of XLH. Burosumab was well tolerated and was effective in improving phosphate metabolism, bone health, and PROs. All the benefits of burosumab were lost after its discontinuation. These results suggested that continuing burosumab is required to achieve and maintain the clinical benefits of the treatment during the transition to young adulthood in patients with XLH.


Subject(s)
Antibodies, Monoclonal, Humanized , Familial Hypophosphatemic Rickets , Fibroblast Growth Factor-23 , Phosphates , Quality of Life , Humans , Familial Hypophosphatemic Rickets/drug therapy , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/pathology , Adolescent , Male , Phosphates/blood , Antibodies, Monoclonal, Humanized/therapeutic use , Female , Child , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/blood
18.
J Med Chem ; 67(14): 11769-11788, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39013015

ABSTRACT

Here, we describe the development of the FGF21 analog zalfermin (NNC0194-0499, 15), intended for once-weekly sc dosing. Protein engineering was needed to address inherent druggability issues of the natural FGF21 hormone. Thus, deamidation of Asp121 was solved by mutation to glutamine, and oxidation of Met168 was solved by mutation to leucine. N-terminal region degradation by dipeptidyl peptidase IV was prevented by alanine residue elongation. To prevent inactivating metabolism by fibroblast activation protein and carboxypeptidase-like activity in the C-terminal region, and to achieve t1/2 extension (53 h in cynomolgus monkeys), we introduced a C18 fatty diacid at the penultimate position 180. The fatty diacid binds albumin in a reversible manner, such that the free fraction of zalfermin potently activates the FGF-receptor complex and retains receptor selectivity compared with FGF21, providing strong efficacy on body weight loss in diet-induced obese mice. Zalfermin is currently being clinically evaluated for the treatment of metabolic dysfunction-associated steatohepatitis.


Subject(s)
Fibroblast Growth Factors , Macaca fascicularis , Fibroblast Growth Factors/metabolism , Animals , Mice , Humans , Male , Proteolysis/drug effects , Mice, Obese , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism
19.
ASN Neuro ; 16(1): 2371163, 2024.
Article in English | MEDLINE | ID: mdl-39024549

ABSTRACT

The fifteen canonical paracrine fibroblast growth factors (FGFs) are organized in five subfamilies that interact with four FGF-receptors (FGFRs) and heparan sulfate proteoglycan (HSPG) co-receptors. Many of these FGFs are expressed in CNS regions where oligodendrocyte (OL) progenitors originate, migrate or differentiate. FGF2 (basic FGF) is considered a prototype FGF and the information about the effects of FGF signaling on OL-lineage cells has evolved largely from the study of FGF2. However, other FGFs from four subfamilies ((FGF1 (FGF1,-2), FGF4 (FGF4,-5,-6), FGF8 (FGF8,-17,-18) and FGF9 (FGF9,-16,-20)) that can interact with the isoforms of FGFRs expressed in OL-lineage cells may also play important roles. We previously reported OL-responses to FGF8 family members. Here, we investigate the effects of members of the FGF1,-4, and -9 subfamilies on proliferation and differentiation of OL progenitors (OPCs), and on cell cycle re-entry and down-regulation of myelin proteins by mature OLs. We found that while FGF2 induced all these responses strongly, FGF4,-6,-9 could do so only transiently and in the presence of exogenous HSPGs, and that FGF5,-16,-20 could not do so even in the presence of heparin or at higher concentrations. Furthermore, we noted that structurally similar FGFs within subfamilies did not always show similarities in their biological effects on OL-lineage cells. Taken together, these studies reveal that FGFs differ in the way they regulate the OL-lineage cells, emphasizes the selectivity and importance of HSPGs as FGF co-receptors in OL-lineage cells and suggests that structural similarity among FGF-subfamily members may not always predict their overlapping biological functions.


Structurally similar members of the FGF1, -4, and -9 subfamilies trigger diverse biological responses in oligodendrocyte-lineage cells and exhibit selective requirement for heparan sulfate proteoglycans as FGF co-receptors.


Subject(s)
Cell Differentiation , Fibroblast Growth Factors , Oligodendroglia , Animals , Oligodendroglia/metabolism , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Cell Differentiation/physiology , Cell Differentiation/drug effects , Cell Proliferation/physiology , Cell Proliferation/drug effects , Cells, Cultured , Structure-Activity Relationship , Rats
20.
Cell Rep Med ; 5(7): 101629, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38959886

ABSTRACT

Weight loss is often followed by weight regain. Characterizing endocrine alterations accompanying weight reduction and regain may disentangle the complex biology of weight-loss maintenance. Here, we profile energy-balance-regulating metabokines and sphingolipids in adults with obesity undergoing an initial low-calorie diet-induced weight loss and a subsequent weight-loss maintenance phase with exercise, glucagon-like peptide-1 (GLP-1) analog therapy, both combined, or placebo. We show that circulating growth differentiation factor 15 (GDF15) and C16:0-C18:0 ceramides transiently increase upon initial diet-induced weight loss. Conversely, circulating fibroblast growth factor 21 (FGF21) is downregulated following weight-loss maintenance with combined exercise and GLP-1 analog therapy, coinciding with increased adiponectin, decreased leptin, and overall decrements in ceramide and sphingosine-1-phosphate levels. Subgroup analyses reveal differential alterations in FGF21-adiponectin-leptin-sphingolipids between weight maintainers and regainers. Clinically, cardiometabolic health outcomes associate with selective metabokine-sphingolipid remodeling signatures. Collectively, our findings indicate distinct FGF21, GDF15, and ceramide responses to diverse phases of weight change and suggest that weight-loss maintenance involves alterations within the metabokine-sphingolipid axis.


Subject(s)
Adiponectin , Fibroblast Growth Factors , Leptin , Sphingolipids , Weight Loss , Humans , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/blood , Adiponectin/blood , Adiponectin/metabolism , Leptin/blood , Leptin/metabolism , Sphingolipids/metabolism , Sphingolipids/blood , Male , Female , Obesity/metabolism , Obesity/blood , Middle Aged , Adult , Ceramides/metabolism , Ceramides/blood , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/blood
SELECTION OF CITATIONS
SEARCH DETAIL