Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Children (Basel) ; 10(4)2023 Apr 03.
Article in English | MEDLINE | ID: mdl-37189930

ABSTRACT

The aim of this research was to characterize cognitive abilities in patients with Glut1-Deficiency syndrome (Glut1DS) following ketogenic diet therapy (KDT). METHODS: The cognitive profiles of eight children were assessed using the Wechsler Intelligence Scale (WISC-IV). The effect of ketogenic diet therapy (KDT) on individual subareas of intelligence was analyzed considering the potential influence of speech motor impairments. RESULTS: Patients with Glut1DS showed a wide range of cognitive performance levels. Some participants showed statistically and clinically significant discrepancies between individual subdomains of intelligence. Both variables, KDT initiation as well as duration, had a positive effect on the overall IQ score. Significant correlations were partially found between the time of KDT initiation and the level of IQ scores, depending on the presence of expressive language test demands of the respective subtests of the WISC-IV. Accordingly, the participants benefited les in the linguistic cognitive domain. The discrepancies in cognitive performance profiles of patients with Glut1DS can be attributed to the possibility of a negative distortion of the results due to the influence of speech motor impairments. CONCLUSIONS: The individual access skills of test persons should be more strongly considered in test procedures for the assessment of intelligence to reduce the negative influence of motor deficits on test performance. Specific characterization and systematization of the speech disorder are indispensable for determining the severity of speech motor impairment in Glut1DS. Therefore, a stronger focus on dysarthria during diagnosis and therapy is necessary.

2.
JCI Insight ; 4(17)2019 09 05.
Article in English | MEDLINE | ID: mdl-31484825

ABSTRACT

Dysregulated actions of bone-derived phosphaturic hormone fibroblast growth factor 23 (FGF23) result in several inherited diseases, such as X-linked hypophosphatemia (XLH), and contribute substantially to the mortality in kidney failure. Mechanisms governing FGF23 production are poorly defined. We herein found that ablation of the Gq/11α-like, extralarge Gα subunit (XLαs), a product of GNAS, exhibits FGF23 deficiency and hyperphosphatemia in early postnatal mice (XLKO). FGF23 elevation in response to parathyroid hormone, a stimulator of FGF23 production via cAMP, was intact in XLKO mice, while skeletal levels of protein kinase C isoforms α and δ (PKCα and PKCδ) were diminished. XLαs ablation in osteocyte-like Ocy454 cells suppressed the levels of FGF23 mRNA, inositol 1,4,5-trisphosphate (IP3), and PKCα/PKCδ proteins. PKC activation in vivo via injecting phorbol myristate acetate (PMA) or by constitutively active Gqα-Q209L in osteocytes and osteoblasts promoted FGF23 production. Molecular studies showed that the PKC activation-induced FGF23 elevation was dependent on MAPK signaling. The baseline PKC activity was elevated in bones of Hyp mice, a model of XLH. XLαs ablation significantly, but modestly, reduced serum FGF23 and elevated serum phosphate in Hyp mice. These findings reveal a potentially hitherto-unknown mechanism of FGF23 synthesis involving a G protein-coupled IP3/PKC pathway, which may be targeted to fine-tune FGF23 levels.


Subject(s)
Fibroblast Growth Factors/metabolism , GTP-Binding Proteins/metabolism , Protein Kinase C/metabolism , Animals , Bone and Bones/metabolism , Disease Models, Animal , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/pathology , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Genetic Predisposition to Disease/genetics , Humans , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism , Osteocytes , Parathyroid Hormone/metabolism , Protein Kinases , RNA, Messenger/metabolism , Recombinant Proteins
3.
Sci Rep ; 8(1): 15973, 2018 10 29.
Article in English | MEDLINE | ID: mdl-30374109

ABSTRACT

Secreted FGF binding proteins (FGFBP) mobilize locally-acting paracrine FGFs from their extracellular storage. Here, we report that FGFBP3 (BP3) modulates fat and glucose metabolism in mouse models of metabolic syndrome. BP3 knockout mice exhibited altered lipid metabolism pathways with reduced hepatic and serum triglycerides. In obese mice the expression of exogenous BP3 reduced hyperglycemia, hepatosteatosis and weight gain, blunted de novo lipogenesis in liver and adipose tissues, increased circulating adiponectin and decreased NEFA. The BP3 protein interacts with endocrine FGFs through its C-terminus and thus enhances their signaling. We propose that BP3 may constitute a new therapeutic to reverse the pathology associated with metabolic syndrome that includes nonalcoholic fatty liver disease and type 2 diabetes mellitus.


Subject(s)
Carbohydrate Metabolism/genetics , Carrier Proteins/genetics , Lipid Metabolism/genetics , Metabolic Syndrome/pathology , Adipose Tissue/metabolism , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Gluconeogenesis/genetics , Glucose Tolerance Test , Lipogenesis/genetics , Liver/metabolism , Metabolic Syndrome/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Protein Binding , STAT3 Transcription Factor/metabolism , Signal Transduction , Triglycerides/blood
4.
FASEB J ; 32(7): 3752-3764, 2018 07.
Article in English | MEDLINE | ID: mdl-29481308

ABSTRACT

Severe anemia and iron deficiency are common complications in chronic kidney disease. The cause of renal anemia is multifactorial and includes decreased erythropoietin (Epo) production, iron deficiency, and inflammation, and it is currently treated with injections of synthetic Epo. However, the use of recombinant Epo has several adverse effects. We previously reported that high fibroblast growth factor 23 (FGF23) levels in mice are associated with decreased red blood cell production, whereas genetic inactivation of Fgf23 results in expansion of the erythroid lineage. The present study is the first to show that high FGF23 levels in a mouse model of renal failure contribute to renal anemia, and inhibiting FGF23 signaling stimulates erythropoiesis and abolishes anemia and iron deficiency. Moreover, we show that inhibition of FGF23 signaling significantly decreases erythroid cell apoptosis and influences the commitment of hematopoietic stem cells toward the erythroid linage. Furthermore, we show that blocking FGF23 signaling attenuates inflammation, resulting in increased serum iron and ferritin levels. Our data clearly demonstrate that elevated FGF23 is a causative factor in the development of renal anemia and iron deficiency, and importantly, blocking FGF23 signaling represents a novel approach to stimulate erythropoiesis and possibly improve survival for millions of chronic kidney disease patients worldwide.-Agoro, R., Montagna, A., Goetz, R., Aligbe, O., Singh, G., Coe, L. M., Mohammadi, M., Rivella, S., Sitara, D. Inhibition of fibroblast growth factor 23 (FGF23) signaling rescues renal anemia.


Subject(s)
Anemia, Iron-Deficiency/metabolism , Fibroblast Growth Factors/antagonists & inhibitors , Renal Insufficiency, Chronic/complications , Signal Transduction , Anemia, Iron-Deficiency/drug therapy , Animals , Apoptosis , Cells, Cultured , Erythroid Cells/metabolism , Ferritins/blood , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Iron/blood , Male , Mice , Mice, Inbred C57BL , Oligopeptides/pharmacology , Oligopeptides/therapeutic use
5.
Nature ; 553(7689): 461-466, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29342138

ABSTRACT

The ageing suppressor α-klotho binds to the fibroblast growth factor receptor (FGFR). This commits FGFR to respond to FGF23, a key hormone in the regulation of mineral ion and vitamin D homeostasis. The role and mechanism of this co-receptor are unknown. Here we present the atomic structure of a 1:1:1 ternary complex that consists of the shed extracellular domain of α-klotho, the FGFR1c ligand-binding domain, and FGF23. In this complex, α-klotho simultaneously tethers FGFR1c by its D3 domain and FGF23 by its C-terminal tail, thus implementing FGF23-FGFR1c proximity and conferring stability. Dimerization of the stabilized ternary complexes and receptor activation remain dependent on the binding of heparan sulfate, a mandatory cofactor of paracrine FGF signalling. The structure of α-klotho is incompatible with its purported glycosidase activity. Thus, shed α-klotho functions as an on-demand non-enzymatic scaffold protein that promotes FGF23 signalling.


Subject(s)
Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Glucuronidase/chemistry , Glucuronidase/metabolism , Paracrine Communication , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , Animals , Binding Sites/genetics , Body Fluids/metabolism , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Glucuronidase/genetics , Heparitin Sulfate/metabolism , Humans , Klotho Proteins , Ligands , Male , Mice , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Protein Binding , Protein Domains , Protein Multimerization , Solubility
7.
Nephrol Dial Transplant ; 30(2): 223-33, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25324355

ABSTRACT

BACKGROUND: αKlotho is the prototypic member of the Klotho family and is most highly expressed in the kidney. αKlotho has pleiotropic biologic effects, and in the kidney, its actions include regulation of ion transport, cytoprotection, anti-oxidation and anti-fibrosis. In rodent models of chronic kidney disease (CKD), αKlotho deficiency has been shown to be an early biomarker as well as a pathogenic factor. The database for αKlotho in human CKD remains controversial even after years of study. METHODS: We used a synthetic antibody library to identify a high-affinity human antigen-binding fragment that recognizes human, rat and mouse αKlotho primarily in its native, rather than denatured, form. RESULTS: Using an immunoprecipitation-immunoblot (IP-IB) assay, we measured both serum and urinary levels of full-length soluble αKlotho in humans and established that human CKD is associated with αKlotho deficiency in serum and urine. αKlotho levels were detectably lower in early CKD preceding disturbances in other parameters of mineral metabolism and progressively declined with CKD stages. We also found that exogenously added αKlotho is inherently unstable in the CKD milieu suggesting that decreased production may not be the sole reason for αKlotho deficiency. CONCLUSION: Synthetic antibody libraries harbor tremendous potential for a variety of biomedical and clinical applications. Using such a reagent, we furnish data in support of αKlotho deficiency in human CKD, and we set the foundation for the development of diagnostic and therapeutic applications of anti-αKlotho antibodies.


Subject(s)
Antibodies/blood , Glucuronidase/deficiency , Renal Insufficiency, Chronic/enzymology , Animals , Biomarkers/blood , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factors/blood , Glucuronidase/immunology , Healthy Volunteers , Humans , Immunoblotting , Immunoglobulin Fab Fragments/blood , Immunoprecipitation , Klotho Proteins , Mice , Peptide Library , Rats
8.
Nature ; 513(7518): 436-9, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25043058

ABSTRACT

Fibroblast growth factor 1 (FGF1) is an autocrine/paracrine regulator whose binding to heparan sulphate proteoglycans effectively precludes its circulation. Although FGF1 is known as a mitogenic factor, FGF1 knockout mice develop insulin resistance when stressed by a high-fat diet, suggesting a potential role in nutrient homeostasis. Here we show that parenteral delivery of a single dose of recombinant FGF1 (rFGF1) results in potent, insulin-dependent lowering of glucose levels in diabetic mice that is dose-dependent but does not lead to hypoglycaemia. Chronic pharmacological treatment with rFGF1 increases insulin-dependent glucose uptake in skeletal muscle and suppresses the hepatic production of glucose to achieve whole-body insulin sensitization. The sustained glucose lowering and insulin sensitization attributed to rFGF1 are not accompanied by the side effects of weight gain, liver steatosis and bone loss associated with current insulin-sensitizing therapies. We also show that the glucose-lowering activity of FGF1 can be dissociated from its mitogenic activity and is mediated predominantly via FGF receptor 1 signalling. Thus we have uncovered an unexpected, neomorphic insulin-sensitizing action for exogenous non-mitogenic human FGF1 with therapeutic potential for the treatment of insulin resistance and type 2 diabetes.


Subject(s)
Fibroblast Growth Factor 1/pharmacology , Glucose/metabolism , Insulin/metabolism , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Dose-Response Relationship, Drug , Fibroblast Growth Factor 1/administration & dosage , Fibroblast Growth Factor 1/adverse effects , Glucose Tolerance Test , Humans , Insulin Resistance , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mitogens/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism
9.
EMBO J ; 33(3): 229-46, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24434184

ABSTRACT

αKlotho is thought to activate the epithelial calcium channel Transient Receptor Potential Vanilloid-5 (TRPV5) in distal renal tubules through its putative glucuronidase/sialidase activity, thereby preventing renal calcium loss. However, αKlotho also functions as the obligatory co-receptor for fibroblast growth factor-23 (FGF23), a bone-derived phosphaturic hormone. Here, we show that renal calcium reabsorption and renal membrane abundance of TRPV5 are reduced in Fgf23 knockout mice, similar to what is seen in αKlotho knockout mice. We further demonstrate that αKlotho neither co-localizes with TRPV5 nor is regulated by FGF23. Rather, apical membrane abundance of TRPV5 in renal distal tubules and thus renal calcium reabsorption are regulated by FGF23, which binds the FGF receptor-αKlotho complex and activates a signaling cascade involving ERK1/2, SGK1, and WNK4. Our data thereby identify FGF23, not αKlotho, as a calcium-conserving hormone in the kidney.


Subject(s)
Calcium/metabolism , Fibroblast Growth Factors/metabolism , Kidney/metabolism , Receptors, Cell Surface/metabolism , TRPV Cation Channels/metabolism , Animals , Cell Membrane/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Glucuronidase , Immediate-Early Proteins/metabolism , Klotho Proteins , Male , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
10.
PLoS Genet ; 9(12): e1003975, 2013.
Article in English | MEDLINE | ID: mdl-24348262

ABSTRACT

Klotho acts as a co-receptor for and dictates tissue specificity of circulating FGF23. FGF23 inhibits PTH secretion, and reduced Klotho abundance is considered a pathogenic factor in renal secondary hyperparathyroidism. To dissect the role of parathyroid gland resident Klotho in health and disease, we generated mice with a parathyroid-specific Klotho deletion (PTH-KL(-/-)). PTH-KL(-/-) mice had a normal gross phenotype and survival; normal serum PTH and calcium; unaltered expression of the PTH gene in parathyroid tissue; and preserved PTH response and sensitivity to acute changes in serum calcium. Their PTH response to intravenous FGF23 delivery or renal failure did not differ compared to their wild-type littermates despite disrupted FGF23-induced activation of the MAPK/ERK pathway. Importantly, calcineurin-NFAT signaling, defined by increased MCIP1 level and nuclear localization of NFATC2, was constitutively activated in PTH-KL(-/-) mice. Treatment with the calcineurin-inhibitor cyclosporine A abolished FGF23-mediated PTH suppression in PTH-KL(-/-) mice whereas wild-type mice remained responsive. Similar results were observed in thyro-parathyroid explants ex vivo. Collectively, we present genetic and functional evidence for a novel, Klotho-independent, calcineurin-mediated FGF23 signaling pathway in parathyroid glands that mediates suppression of PTH. The presence of Klotho-independent FGF23 effects in a Klotho-expressing target organ represents a paradigm shift in the conceptualization of FGF23 endocrine action.


Subject(s)
Fibroblast Growth Factors/genetics , Membrane Proteins/genetics , Parathyroid Hormone/genetics , Signal Transduction/genetics , Animals , Calcineurin/metabolism , Calcineurin Inhibitors , Calcium/blood , Cyclosporine/pharmacology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Gene Expression Regulation/drug effects , Humans , Kidney/metabolism , Klotho Proteins , Membrane Proteins/metabolism , Mice , Parathyroid Glands/metabolism , Parathyroid Hormone/blood , Sequence Deletion , Vitamin D/metabolism
12.
Contrib Nephrol ; 180: 124-37, 2013.
Article in English | MEDLINE | ID: mdl-23652555

ABSTRACT

Deregulated phosphate homeostasis can lead to a wide range of disorders, including myopathy, cardiac dysfunction, and skeletal abnormalities. Therefore, characterization of the molecular regulation of phosphate metabolism is of pathophysiological and clinical significance. Hyp mouse is the model for human X-linked hypophosphatemia which is due to mutations that inactivate the endopeptidases of the X chromosome (PHEX). PHEX inactivation leads to increased serum levels of fibroblast growth factor 23 (FGF23), a phosphaturic hormone that induces excessive renal phosphate excretion and severe hypophosphatemia. The expression of WNT signaling components is increased in Hyp mice. To determine the potential role of WNT signaling in FGF23-mediated hypophosphatemia, we cross-bred Hyp mice with mice deficient in the WNT coreceptor low-density lipoprotein receptor-related protein 6 (Lrp6) to generate Hyp and Lrp6 double mutant mice (Hyp/Lrp6). Like Hyp mice, Hyp/Lrp6 double mutants maintained high serum levels of FGF23, and accordingly exhibited hypophosphatemia to the same degree as the Hyp mice did, indicating that genetically reducing WNT signaling does not impact FGF23-induced phosphaturia. Moreover, similar to Hyp mice, the Hyp/Lrp6 double mutants also exhibited reduced mineralization of the bone, further supporting that reduced WNT signaling does not affect the chronic phosphate wasting caused by excess FGF23 in these mice. In further support of our finding, injection of bioactive FGF23 protein into Lrp6 mutant mice reduced serum phosphate levels to a similar degree as FGF23 injection into wild-type mice. Our in vivo studies provide genetic and pharmacological evidence for a WNT-independent function of FGF23 in the regulation of phosphate homeostasis.


Subject(s)
Disease Models, Animal , Familial Hypophosphatemic Rickets/physiopathology , Fibroblast Growth Factors/physiology , Low Density Lipoprotein Receptor-Related Protein-6/deficiency , PHEX Phosphate Regulating Neutral Endopeptidase/physiology , Wnt Signaling Pathway , Animals , Familial Hypophosphatemic Rickets/diagnostic imaging , Familial Hypophosphatemic Rickets/etiology , Familial Hypophosphatemic Rickets/genetics , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/toxicity , Homeostasis , Hypophosphatemia, Familial/genetics , Hypophosphatemia, Familial/metabolism , Kidney/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/physiology , Male , Mice , Mice, Knockout , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Phosphates/metabolism , Radiography , Recombinant Proteins/toxicity , Sodium-Phosphate Cotransporter Proteins, Type II/biosynthesis , Sodium-Phosphate Cotransporter Proteins, Type II/genetics
13.
PLoS One ; 8(4): e60658, 2013.
Article in English | MEDLINE | ID: mdl-23577141

ABSTRACT

Recent studies support a role for FGF23 and its co-receptor Klotho in cardiovascular pathology, yet the underlying mechanisms remain largely elusive. Herein, we analyzed the expression of Klotho in mouse arteries and generated a novel mouse model harboring a vascular smooth muscle cell specific deletion of Klotho (Sm22-KL(-/-) ). Arterial Klotho expression was detected at very low levels with quantitative real-time PCR; Klotho protein levels were undetectable by immunohistochemistry and Western blot. There was no difference in arterial Klotho between Sm22-KL(-/-) and wild-type mice, as well as no changes in serum markers of mineral metabolism. Intravenous delivery of FGF23 elicited a rise in renal (0.005; p<0.01) but not arterial Egr-1 expression, a marker of Klotho-dependent FGF23 signaling. Further, the impact of FGF23 on vascular calcification and endothelial response was evaluated in bovine vascular smooth muscle cells (bVSMC) and in a murine ex vivo model of endothelial function, respectively. FGF23 treatment (0.125-2 ng/mL) did not modify calcification in bVSMCs or dilatory, contractile and structural properties in mice arterial specimen ex vivo. Collectively, these results demonstrate that FGF23-Klotho signaling is absent in mouse arteries and that the vascular response was unaffected by FGF23 treatment. Thus, our data do not support Klotho-mediated FGF23 effects in the vasculature although confirmative studies in humans are warranted.


Subject(s)
Arteries/drug effects , Arteries/metabolism , Calcinosis/metabolism , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation/drug effects , Glucuronidase/metabolism , Animals , Arteries/pathology , Arteries/physiopathology , Calcinosis/genetics , Cattle , Early Growth Response Protein 1/genetics , Female , Fibroblast Growth Factor-23 , Gene Deletion , Glucuronidase/deficiency , Glucuronidase/genetics , Klotho Proteins , Male , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Phenotype , Time Factors
14.
Nat Rev Mol Cell Biol ; 14(3): 166-80, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23403721

ABSTRACT

Fibroblast growth factors (FGFs) mediate a broad range of functions in both the developing and adult organism. The accumulated wealth of structural information on the FGF signalling pathway has begun to unveil the underlying molecular mechanisms that modulate this system to generate a myriad of distinct biological outputs in development, tissue homeostasis and metabolism. At the ligand and receptor level, these mechanisms include alternative splicing of the ligand (FGF8 subfamily) and the receptor (FGFR1-FGFR3), ligand homodimerization (FGF9 subfamily), site-specific proteolytic cleavage of the ligand (FGF23), and interaction of the ligand and the receptor with heparan sulphate cofactor and Klotho co-receptor.


Subject(s)
Fibroblast Growth Factors/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Alternative Splicing , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Fibroblast Growth Factor 9/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/chemistry , Glucuronidase/metabolism , Heparitin Sulfate/metabolism , Klotho Proteins , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism
15.
Bone ; 51(3): 621-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22647968

ABSTRACT

Fibroblast growth factor-23 (FGF23) is a bone-derived endocrine regulator of phosphate homeostasis which inhibits renal tubular phosphate reabsorption. Binding of circulating FGF23 to FGF receptors in the cell membrane requires the concurrent presence of the co-receptor αKlotho. It is still controversial whether αKlotho is expressed in the kidney proximal tubule, the principal site of phosphate reabsorption. Hence, it has remained an enigma as to how FGF23 downregulates renal phosphate reabsorption. Here, we show that renal proximal tubular cells do express the co-receptor αKlotho together with cognate FGF receptors, and that FGF23 directly downregulates membrane expression of the sodium-phosphate cotransporter NaPi-2a by serine phosphorylation of the scaffolding protein Na(+)/H(+) exchange regulatory cofactor (NHERF)-1 through ERK1/2 and serum/glucocorticoid-regulated kinase-1 signaling.


Subject(s)
Fibroblast Growth Factors/pharmacology , Hypophosphatemia, Familial/enzymology , Hypophosphatemia, Familial/pathology , Immediate-Early Proteins/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/enzymology , MAP Kinase Signaling System/drug effects , Protein Serine-Threonine Kinases/metabolism , Animals , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/pathology , Fibroblast Growth Factor-23 , Gene Expression Regulation/drug effects , Glucuronidase/genetics , Glucuronidase/metabolism , Humans , Immediate-Early Proteins/genetics , Kidney Tubules, Proximal/pathology , Klotho Proteins , Male , Mice , Mice, Inbred C57BL , Models, Biological , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Sodium-Hydrogen Exchangers/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism
16.
J Biol Chem ; 287(34): 29134-46, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22733815

ABSTRACT

FGFs 19, 21, and 23 are hormones that regulate in a Klotho co-receptor-dependent fashion major metabolic processes such as glucose and lipid metabolism (FGF21) and phosphate and vitamin D homeostasis (FGF23). The role of heparan sulfate glycosaminoglycan in the formation of the cell surface signaling complex of endocrine FGFs has remained unclear. Here we show that heparan sulfate is not a component of the signal transduction unit of FGF19 and FGF23. In support of our model, we convert a paracrine FGF into an endocrine ligand by diminishing heparan sulfate-binding affinity of the paracrine FGF and substituting its C-terminal tail for that of an endocrine FGF containing the Klotho co-receptor-binding site to home the ligand into the target tissue. In addition to serving as a proof of concept, the ligand conversion provides a novel strategy for engineering endocrine FGF-like molecules for the treatment of metabolic disorders, including global epidemics such as type 2 diabetes and obesity.


Subject(s)
Fibroblast Growth Factors/metabolism , Heparitin Sulfate/metabolism , Models, Biological , Paracrine Communication , Signal Transduction , Animals , Cell Line, Tumor , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Endocrine System/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Heparitin Sulfate/genetics , Humans , Mice , Mice, Mutant Strains , Obesity/genetics , Obesity/metabolism , Obesity/therapy
17.
Mol Cell Biol ; 32(10): 1944-54, 2012 May.
Article in English | MEDLINE | ID: mdl-22451487

ABSTRACT

It has been recently established that Klotho coreceptors associate with fibroblast growth factor (FGF) receptor tyrosine kinases (FGFRs) to enable signaling by endocrine-acting FGFs. However, the molecular interactions leading to FGF-FGFR-Klotho ternary complex formation remain incompletely understood. Here, we show that in contrast to αKlotho, ßKlotho binds its cognate endocrine FGF ligand (FGF19 or FGF21) and FGFR independently through two distinct binding sites. FGF19 and FGF21 use their respective C-terminal tails to bind to a common binding site on ßKlotho. Importantly, we also show that Klotho coreceptors engage a conserved hydrophobic groove in the immunoglobulin-like domain III (D3) of the "c" splice isoform of FGFR. Intriguingly, this hydrophobic groove is also used by ligands of the paracrine-acting FGF8 subfamily for receptor binding. Based on this binding site overlap, we conclude that while Klotho coreceptors enhance binding affinity of FGFR for endocrine FGFs, they actively suppress binding of FGF8 subfamily ligands to FGFR.


Subject(s)
Fibroblast Growth Factor 8/metabolism , Glucuronidase/metabolism , Paracrine Communication , Signal Transduction , Animals , Binding Sites , Cell Line , Humans , Klotho Proteins , Ligands , Protein Isoforms/metabolism , Rats , Receptors, Fibroblast Growth Factor/metabolism
18.
Proc Natl Acad Sci U S A ; 109(8): 3143-8, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22315431

ABSTRACT

The endocrine hormone fibroblast growth factor 21 (FGF21) is a powerful modulator of glucose and lipid metabolism and a promising drug for type 2 diabetes. Here we identify FGF21 as a potent regulator of skeletal homeostasis. Both genetic and pharmacologic FGF21 gain of function lead to a striking decrease in bone mass. In contrast, FGF21 loss of function leads to a reciprocal high-bone-mass phenotype. Mechanistically, FGF21 inhibits osteoblastogenesis and stimulates adipogenesis from bone marrow mesenchymal stem cells by potentiating the activity of peroxisome proliferator-activated receptor γ (PPAR-γ). Consequently, FGF21 deletion prevents the deleterious bone loss side effect of the PPAR-γ agonist rosiglitazone. Therefore, FGF21 is a critical rheostat for bone turnover and a key integrator of bone and energy metabolism. These results reveal that skeletal fragility may be an undesirable consequence of chronic FGF21 administration.


Subject(s)
Bone Resorption/pathology , Fibroblast Growth Factors/metabolism , PPAR gamma/metabolism , Adipogenesis/drug effects , Animals , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Resorption/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Drug Resistance/drug effects , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/pharmacology , Humans , Mice , Mice, Knockout , Organ Size/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteogenesis/drug effects , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Rosiglitazone , Thiazolidinediones/pharmacology
19.
Structure ; 20(1): 77-88, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22244757

ABSTRACT

Uncontrolled fibroblast growth factor (FGF) signaling can lead to human malignancies necessitating multiple layers of self-regulatory control mechanisms. Fibroblast growth factor receptor (FGFR) autoinhibition mediated by the alternatively spliced immunoglobulin (Ig) domain 1 (D1) and the acid box (AB)-containing linker between D1 and Ig domain 2 (D2) serves as the first line of defense to minimize inadvertent FGF signaling. In this report, nuclear magnetic resonance and surface plasmon resonance spectroscopy are used to demonstrate that the AB subregion of FGFR electrostatically engages the heparan sulfate (HS)-binding site on the D2 domain in cis to directly suppress HS-binding affinity of FGFR. Furthermore, the cis electrostatic interaction sterically autoinhibits ligand-binding affinity of FGFR because of the close proximity of HS-binding and primary ligand-binding sites on the D2 domain. These data, together with the strong amino acid sequence conservation of the AB subregion among FGFR orthologs, highlight the universal role of the AB subregion in FGFR autoinhibition.


Subject(s)
Fibroblast Growth Factors/metabolism , Models, Molecular , Protein Structure, Tertiary/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/physiology , Alternative Splicing/genetics , Amino Acid Sequence , Conserved Sequence , Heparitin Sulfate/metabolism , Humans , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding/genetics , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction/genetics , Static Electricity , Surface Plasmon Resonance
20.
J Clin Invest ; 121(8): 3220-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21747170

ABSTRACT

The nuclear receptor pregnane X receptor (PXR) is activated by a range of xenochemicals, including chemotherapeutic drugs, and has been suggested to play a role in the development of tumor cell resistance to anticancer drugs. PXR also has been implicated as a regulator of the growth and apoptosis of colon tumors. Here, we have used a xenograft model of colon cancer to define a molecular mechanism that might underlie PXR-driven colon tumor growth and malignancy. Activation of PXR was found to be sufficient to enhance the neoplastic characteristics, including cell growth, invasion, and metastasis, of both human colon tumor cell lines and primary human colon cancer tissue xenografted into immunodeficient mice. Furthermore, we were able to show that this PXR-mediated phenotype required FGF19 signaling. PXR bound to the FGF19 promoter in both human colon tumor cells and "normal" intestinal crypt cells. However, while both cell types proliferated in response to PXR ligands, the FGF19 promoter was activated by PXR only in cancer cells. Taken together, these data indicate that colon cancer growth in the presence of a specific PXR ligand results from tumor-specific induction of FGF19. These observations may lead to improved therapeutic regimens for colon carcinomas.


Subject(s)
Fibroblast Growth Factors/metabolism , Receptors, Steroid/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Caco-2 Cells , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colonic Neoplasms/metabolism , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Phenotype , Plasmids/metabolism , Pregnane X Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...