Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Pediatr Nephrol ; 35(10): 1843-1854, 2020 10.
Article in English | MEDLINE | ID: mdl-31392510

ABSTRACT

Hypophosphatemic rickets is caused by renal phosphate wasting that is most commonly due to X-linked dominant mutations in PHEX. PHEX mutations cause hypophosphatemia indirectly, through the increased expression of fibroblast growth factor 23 (FGF23) by osteocytes. FGF23 decreases renal phosphate reabsorption and thereby increases phosphate excretion. The lack of phosphate leads to a mineralization defect at the level of growth plates (rickets), bone tissue (osteomalacia), and teeth, where the defect facilitates the formation of abscesses. The bone tissue immediately adjacent to osteocytes often remains unmineralized ("periosteocytic lesions"), highlighting the osteocyte defect in this disorder. Common clinical features of XLH include deformities of the lower extremities, short stature, enthesopathies, dental abscesses, as well as skull abnormalities such as craniosynostosis and Chiari I malformation. For the past four decades, XLH has been treated by oral phosphate supplementation and calcitriol, which improves rickets and osteomalacia and the dental manifestations, but often does not resolve all aspects of the mineralization defects. A newer treatment approach using inactivating FGF23 antibodies leads to more stable control of serum inorganic phosphorus levels and seems to heal rickets more reliably. However, the long-term benefits of FGF23 antibody treatment remain to be elucidated.


Subject(s)
Familial Hypophosphatemic Rickets/pathology , Fibroblast Growth Factors/metabolism , Osteomalacia/pathology , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Phosphates/metabolism , Absorptiometry, Photon , Bone Development/drug effects , Bone Development/genetics , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Calcitriol/administration & dosage , Familial Hypophosphatemic Rickets/diagnosis , Familial Hypophosphatemic Rickets/drug therapy , Familial Hypophosphatemic Rickets/genetics , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/antagonists & inhibitors , Humans , Osteocytes/metabolism , Osteomalacia/diagnosis , Osteomalacia/drug therapy , Osteomalacia/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Paracrine Communication/genetics , Phosphates/administration & dosage , Phosphates/blood , Renal Reabsorption/drug effects , Renal Reabsorption/genetics , Tooth/growth & development , Tooth/pathology , Treatment Outcome
2.
Kidney Int ; 94(1): 49-59, 2018 07.
Article in English | MEDLINE | ID: mdl-29735309

ABSTRACT

High circulating fibroblast growth factor 23 (FGF23) levels are probably a major risk factor for cardiovascular disease in chronic kidney disease. FGF23 interacts with the receptor FGFR4 in cardiomyocytes inducing left ventricular hypertrophy. Moreover, in the liver FGF23 via FGFR4 increases the risk of inflammation which is also found in chronic kidney disease. In contrast, X-linked hypophosphatemia is characterized by high FGF23 circulating levels due to loss of function mutations of the phosphate-regulating gene with homologies to an endopeptidase on the X chromosome (PHEX), but is not characterized by high cardiovascular morbidity. Here we used a novel murine X-linked hypophosphatemia model, the PhexC733RMhda mouse line, bearing an amino acid substitution (p.Cys733Arg) to test whether high circulating FGF23 in the absence of renal injury would trigger cardiovascular disease. As X-linked hypophosphatemia patient mimics, these mice show high FGF23 levels, hypophosphatemia, normocalcemia, and low/normal vitamin D levels. Moreover, these mice show hyperparathyroidism and low circulating soluble αKlotho levels. At the age of 27 weeks we found no left ventricular hypertrophy and no alteration of cardiac function as assessed by echocardiography. These mice also showed no activation of the calcineurin/NFAT pathway in heart and liver and no tissue and systemic signs of inflammation. Importantly, blood pressure, glomerular filtration rate and urea clearance were similar between genotypes. Thus, the presence of high circulating FGF23 levels alone in the absence of renal impairment and normal/high phosphate levels is not sufficient to cause cardiovascular disease.


Subject(s)
Familial Hypophosphatemic Rickets/blood , Fibroblast Growth Factors/blood , Hypertrophy, Left Ventricular/epidemiology , Animals , Disease Models, Animal , Echocardiography , Familial Hypophosphatemic Rickets/genetics , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Heart/diagnostic imaging , Humans , Hypertrophy, Left Ventricular/blood , Hypertrophy, Left Ventricular/diagnosis , Hypertrophy, Left Ventricular/etiology , Loss of Function Mutation , Male , Mice , Mice, Transgenic , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Phosphates/blood , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Risk Factors , X-Ray Microtomography
3.
Hum Mol Genet ; 25(13): 2661-2671, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27126636

ABSTRACT

X-linked hypophosphatemia (XLH) is the most common cause of inheritable rickets, with an incidence of 1/20 000 in humans. Inactivation or mutation of the gene PHEX, a phosphate-regulating endopeptidase, leads to hypophosphatemia and defective bone mineralization in XLH patients. Presently, there is no adequate animal model for safety assessments of physiotherapies and drug screening for XLH rickets. In this study, an XLH model was generated via PHEX gene knockout (KO) through coinjection of clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9)/sgRNA mRNA into rabbit zygotes. The typical phenotypes of growth retardation, hypophosphatemia, elevated serum FGF23 and bone mineralization were observed in the PHEX KO rabbits but not in normal controls. In summary, for the first time, we have successfully obtained PHEX KO rabbits and recapitulated human XLH using the CRISPR/Cas9 system. This novel XLH rabbit model could be utilized as a drug screening model for XLH prevention and preclinical therapy.


Subject(s)
Disease Models, Animal , Familial Hypophosphatemic Rickets/genetics , Animals , Calcification, Physiologic/genetics , Familial Hypophosphatemic Rickets/metabolism , Female , Fibroblast Growth Factor-23 , Gene Knockout Techniques/methods , Gene Knockout Techniques/veterinary , Genetic Diseases, X-Linked/genetics , Humans , Hypophosphatemia/etiology , Hypophosphatemia/genetics , Mutation , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Phosphates , Rabbits , Rickets/etiology
4.
Connect Tissue Res ; 59(sup1): 91-96, 2018 12.
Article in English | MEDLINE | ID: mdl-29745817

ABSTRACT

X-linked hypophosphatemia (XLH) is a skeletal disorder arising from mutations in the PHEX gene, transmitted in most cases as an X-linked dominant trait. PHEX deficiency leads to renal phosphate wasting and hypophosphatemia, as well as impaired mineralization of bone and dentin, resulting in severe skeletal and dental complications. Dentin mineralization defects appear as characteristic, large interglobular spaces resulting from the lack of fusion of calculospherites in the circumpulpal region during the mineralization process. Here, we examined changes in the composition and structure of dentin using Raman spectroscopy on XLH human teeth, and using transmission electron microscopy on the dentin of Hyp mice (the murine model of XLH). The dentin of patients with XLH showed changes in the quality of the apatitic mineral, with greater carbonate substitution and lower crystallinity compared to the dentin of age-matched control teeth. In addition, ultrastructural analysis by transmission electron microscopy revealed a major disorganization of the peri- and intertubular structure of the dentin, with odontoblast processes residing within an unmineralized matrix sheath in the Hyp mouse. Taken together, these results indicate that like for bone and tooth cementum, there are impaired mineral quality and matrix changes in XLH dentin reflecting high sensitivity to systemic serum phosphate levels and possibly other local changes in the dentin matrix.


Subject(s)
Calcification, Physiologic/genetics , Dentin/metabolism , Familial Hypophosphatemic Rickets/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Animals , Dentin/pathology , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/pathology , Female , Humans , Male , Mice , Mice, Mutant Strains , Mutation , PHEX Phosphate Regulating Neutral Endopeptidase/genetics
5.
FASEB J ; 30(1): 121-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26324849

ABSTRACT

Recent studies have identified family with sequence similarity member 20C (FAM20C) as a kinase that phosphorylates the Ser in Ser-X-Glu/phospho-Ser (pSer) motifs in the small-integrin-binding ligand N-linked glycoproteins (SIBLINGs). There is no in vivo evidence that validates this finding, and it is unclear whether FAM20C is the only kinase for SIBLINGs. We extracted bone noncollagenous proteins (NCPs) from Fam20C-knockout (KO) mice and analyzed the phosphorylation levels. The total NCPs were separated into osteopontin-, bone sialoprotein-, and dentin matrix protein-1-enriched fractions by anion-exchange chromatography and analyzed by SDS-PAGE, native PAGE, and Western immunoblot analysis. The NCP phosphorylation level in the KO mice was lower than that in the wild-type (WT). On the native gel, the SIBLINGs from KO mice showed a lower migration rate (Mr) than those from the WT. Calf intestine phosphatase treatment shifted SIBLINGs from the WT mice to the level adjacent to the KO, but failed to shift the latter, suggesting a phosphorylation loss of SIBLINGs in the KO mice. Mass spectrometry identified less pSers in the SIBLINGs from the KO mice [including the region of the acidic Ser- and aspartate-rich motif (ASARM) peptides]. In an intriguing finding, several pSers in the Ser-X-Glu motifs in the KO mice maintained their phosphorylation, whereas several others in non-Ser-X-Glu motifs did not. Phospho-Tyrs and phospho-Thrs in the SIBLINGs did not appear to be associated with FAM20C. Our results indicate that FAM20C is the primary, but not the only, kinase for the SIBLINGs.-Yang, X., Yan, W., Tian, Y., Ma, P., Opperman, L. A., Wang, X. Family with sequence similarity member 20C is the primary but not the only kinase for the small-integrin-binding ligand N-linked glycoproteins in bone.


Subject(s)
Bone and Bones/metabolism , Calcification, Physiologic/physiology , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Integrins/metabolism , Osteopontin/metabolism , Animals , Cattle , Electrophoresis, Polyacrylamide Gel/methods , Mice, Knockout , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism
6.
Virol J ; 14(1): 35, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28222744

ABSTRACT

BACKGROUND: Herpes simplex virus 1 (HSV-1) is an archetypal member of the alphaherpesvirus subfamily with a large genome encoding over 80 proteins, many of which play a critical role in virus-host interactions and immune modulation. Upon viral infections, the host cells activate innate immune responses to restrict their replications. Peroxisomes, which have long been defined to regulate metabolic activities, are reported to be important signaling platforms for antiviral innate immunity. It has been verified that signaling from peroxisomal MAVS (MAVS-Pex) triggers a rapid interferon (IFN) independent IFN-stimulated genes (ISGs) production against invading pathogens. However, little is known about the interaction between DNA viruses such as HSV-1 and the MAVS-Pex mediated signaling. RESULTS: HSV-1 could activate the MAVS-Pex signaling pathway at a low multiplicity of infection (MOI), while infection at a high MOI dampens MAVS-Pex induced immediately early ISGs production. A high-throughput screen assay reveals that HSV-1 tegument protein VP16 inhibits the immediate early ISGs expression downstream of MAVS-Pex signaling. Moreover, the expression of ISGs was recovered when VP16 was knockdown with its specific short hairpin RNA. CONCLUSION: HSV-1 blocks MAVS-Pex mediated early ISGs production through VP16 to dampen the immediate early antiviral innate immunity signaling from peroxisomes.


Subject(s)
Herpes Simplex Virus Protein Vmw65/metabolism , Herpesvirus 1, Human/pathogenicity , Host-Pathogen Interactions , Immune Evasion , Immunity, Innate , Peroxisomes/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Cell Line , Humans , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism
7.
J Biol Regul Homeost Agents ; 31(1): 215-220, 2017.
Article in English | MEDLINE | ID: mdl-28337895

ABSTRACT

Since 1979, Pulsed electromagnetic fields (PEMFs) have been approved by the Food and Drug Administration as an effective method in the treatment of non-unions. As well as PEMFs, also static magnetic fields (SMFs) have been widely investigated in orthopaedic studies. Even if the exact mechanism of action is not well understood, a large number of studies showed specific effects both at cellular and tissue levels. As bone fracture healing and osseointegration share the same biological events, the application of magnetic field stimulation in order to facilitate the osseointegration process has been suggested. In this study we investigated the proliferation rate and gene expression profile of MG63 osteoblastic-like cells after a 24, 48 and 72-hour SMF stimulation, generated by a small, customized cover screw-shaped neodymium-iron-bore magnet placed in the inner cavity of a dental implant. As a result, we found that the application of a SMF to osteoblastic-like cells does slightly decrease cell proliferation rate while enhancing the expression of those genes correlated to differentiation and mineralization. Our findings represent, to our knowledge, the first clinical ready technique for dental implants showing the ability of SMF to promote the osteogenesis process in vitro.


Subject(s)
Bone Regeneration/genetics , Dental Implants , Magnetic Fields , Magnets , Osseointegration/genetics , Osteoblasts/cytology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Biomarkers , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Screws , Cell Differentiation , Cell Line , Cell Proliferation , Collagen Type X/genetics , Collagen Type X/metabolism , Gene Expression , Humans , Osteoblasts/metabolism , Osteogenesis/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
8.
J Inherit Metab Dis ; 39(6): 859-868, 2016 11.
Article in English | MEDLINE | ID: mdl-27469511

ABSTRACT

INTRODUCTION: Zellweger spectrum disorders (ZSDs) are characterized by a failure in peroxisome formation, caused by autosomal recessive mutations in different PEX genes. At least some of the progressive and irreversible clinical abnormalities in patients with a ZSD, particularly liver dysfunction, are likely caused by the accumulation of toxic bile acid intermediates. We investigated whether cholic acid supplementation can suppress bile acid synthesis, reduce accumulation of toxic bile acid intermediates and improve liver function in these patients. METHODS: An open label, pretest-posttest design study was conducted including 19 patients with a ZSD. Participants were followed longitudinally during a period of 2.5 years prior to the start of the intervention. Subsequently, all patients received oral cholic acid and were followed during 9 months of treatment. Bile acids, peroxisomal metabolites, liver function and liver stiffness were measured at baseline and 4, 12 and 36 weeks after start of cholic acid treatment. RESULTS: During cholic acid treatment, bile acid synthesis decreased in the majority of patients. Reduced levels of bile acid intermediates were found in plasma and excretion of bile acid intermediates in urine was diminished. In patients with advanced liver disease (n = 4), cholic acid treatment resulted in increased levels of plasma transaminases, bilirubin and cholic acid with only a minor reduction in bile acid intermediates. CONCLUSIONS: Oral cholic acid therapy can be used in the majority of patients with a ZSD, leading to at least partial suppression of bile acid synthesis. However, caution is needed in patients with advanced liver disease due to possible hepatotoxic effects.


Subject(s)
Cholic Acid/therapeutic use , Zellweger Syndrome/drug therapy , Adolescent , Adult , Bile Acids and Salts/metabolism , Bilirubin/blood , Child , Child, Preschool , Cholic Acid/blood , Female , Humans , Liver/metabolism , Liver Diseases/drug therapy , Liver Diseases/metabolism , Longitudinal Studies , Male , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Transaminases/blood , Young Adult , Zellweger Syndrome/blood , Zellweger Syndrome/metabolism
9.
Am J Physiol Renal Physiol ; 309(9): F764-9, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26336161

ABSTRACT

Nephrogenic systemic fibrosis (NSF) is a devastating condition associated with gadolinium (Gd3+)-based contrast agents (GBCAs) in patients with kidney disease. The release of toxic Gd3+ from GBCAs likely plays a major role in NSF pathophysiology. The cause and etiology of Gd3+ release from GBCAs is unknown. Increased Acidic Serine Aspartate Rich MEPE-associated peptides (ASARM peptides) induce bone mineralization abnormalities and contribute to renal phosphate-handling defects in inherited hypophosphatemic rickets and tumor-induced osteomalacia. The proteolytic cleavage of related bone matrix proteins with ASARM motifs results in release of ASARM peptide into bone and circulation. ASARM peptides are acidic, reactive, phosphorylated inhibitors of mineralization that bind Ca2+ and hydroxyapatite. Since the ionic radius of Gd3+ is close to that of Ca2+, we hypothesized that ASARM peptides increase the risk of NSF by inducing release of Gd3+ from GBCAs. Here, we show 1) ASARM peptides bind and induce release of Gd3+ from GBCAs in vitro and in vivo; 2) A bioengineered peptide (SPR4) stabilizes the Gd3+-GBCA complex by specifically binding to ASARM peptide in vitro and in vivo; and 3) SPR4 peptide infusion prevents GBCA-induced NSF-like pathology in a murine model with increased ASARM peptide (Hyp mouse). We conclude ASARM peptides may play a role in NSF and SPR4 peptide is a candidate adjuvant for preventing or reducing risk of disease.


Subject(s)
Contrast Media , Extracellular Matrix Proteins/metabolism , Gadolinium DTPA , Glycoproteins/metabolism , Kidney/metabolism , Meglumine/analogs & derivatives , Nephrogenic Fibrosing Dermopathy/prevention & control , Organometallic Compounds , PHEX Phosphate Regulating Neutral Endopeptidase/pharmacology , Peptide Fragments/pharmacology , Phosphoproteins/metabolism , Animals , Cytoprotection , Disease Models, Animal , Drug Stability , Familial Hypophosphatemic Rickets/complications , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/metabolism , Fibroblast Growth Factor-23 , Kidney/diagnostic imaging , Kidney/pathology , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Nephrogenic Fibrosing Dermopathy/chemically induced , Nephrogenic Fibrosing Dermopathy/diagnosis , Nephrogenic Fibrosing Dermopathy/genetics , Nephrogenic Fibrosing Dermopathy/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Peptide Fragments/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , X-Ray Microtomography
10.
Biol Chem ; 396(1): 27-33, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25060345

ABSTRACT

Familial hypophosphatemic rickets (HR), the most common inherited form of rickets, is a group of inherited renal phosphate wasting disorders characterized by growth retardation, rickets with bone deformities, osteomalacia, poor dental development, and hypophosphatemia. The purpose of this study was to identify the genetic defect responsible for familial HR in a four-generation Chinese Han pedigree by exome sequencing and Sanger sequencing. Clinical features include skeletal deformities, teeth abnormalities, hearing impairments and variable serum phosphate level in patients of this family. A novel deletion mutation, c.1553delT (p.F518Sfs*4), was identified in the X-linked phosphate regulating endopeptidase homolog gene (PHEX). The mutation is predicted to result in prematurely truncated and loss-of-function PHEX protein. Our data suggest that exome sequencing is a powerful tool to discover mutation(s) in HR, a disorder with genetic and clinical heterogeneity. The findings may also provide new insights into the cause and diagnosis of HR, and have implications for genetic counseling and clinical management.


Subject(s)
Exome/genetics , Familial Hypophosphatemic Rickets/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Adolescent , Adult , Child , China , Female , Genetic Counseling , Humans , Male , Middle Aged , Mutation , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Sequence Analysis
11.
J Transl Med ; 13: 296, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26362198

ABSTRACT

BACKGROUND: Peripheral nerve injury and bone lesions, well known leprosy complications, lead to deformities and incapacities. The phosphate-regulating gene with homologies to endopeptidase on the X chromosome (PHEX) encodes a homonymous protein (PHEX) implicated in bone metabolism. PHEX/PHEX alterations may result in bone and cartilage lesions. PHEX expression is downregulated by intracellular Mycobacterium leprae (M. leprae) in cultures of human Schwann cells and osteoblasts. M. leprae in vivo effect on PHEX/PHEX is not known. METHODS: Cross-sectional observational study of 36 leprosy patients (22 lepromatous and 14 borderline-tuberculoid) and 20 healthy volunteers (HV). The following tests were performed: PHEX flow cytometric analysis on blood mononuclear cells, cytokine production in culture supernatant, 25-hydroxyvitamin D (OHvitD) serum levels and (99m)Tc-MDP three-phase bone scintigraphy, radiography of upper and lower extremities and blood and urine biochemistry. RESULTS: Significantly lower PHEX expression levels were observed in lepromatous patients than in the other groups (χ(2) = 16.554, p < 0.001 for lymphocytes and χ(2) = 13.933, p = 0.001 for monocytes). Low levels of 25-(OHvitD) were observed in HV (median = 23.0 ng/mL) and BT patients (median = 27.5 ng/mL) and normal serum levels were found in LL patients (median = 38.6 ng/mL). Inflammatory cytokines, such as TNF, a PHEX transcription repressor, were lower after stimulation with M. leprae in peripheral blood mononuclear cells from lepromatous in comparison to BT patients and HV (χ(2) = 10.820, p < 0.001). CONCLUSION: Downregulation of PHEX may constitute an important early component of bone loss and joint damage in leprosy. The present results suggest a direct effect produced by M. leprae on the osteoarticular system that may use this mechanism.


Subject(s)
Down-Regulation , Leprosy, Borderline/metabolism , Leprosy, Multibacillary/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Bone and Bones/microbiology , Cartilage/microbiology , Cross-Sectional Studies , Cytokines/metabolism , Female , Flow Cytometry , Healthy Volunteers , Humans , Inflammation/metabolism , Inflammation/microbiology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Osteoblasts/microbiology , Schwann Cells/microbiology , Technetium Tc 99m Medronate , Young Adult
12.
Nat Genet ; 38(11): 1248-50, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17033625

ABSTRACT

Hypophosphatemia is a genetically heterogeneous disease. Here, we mapped an autosomal recessive form (designated ARHP) to chromosome 4q21 and identified homozygous mutations in DMP1 (dentin matrix protein 1), which encodes a non-collagenous bone matrix protein expressed in osteoblasts and osteocytes. Intact plasma levels of the phosphaturic protein FGF23 were clearly elevated in two of four affected individuals, providing a possible explanation for the phosphaturia and inappropriately normal 1,25(OH)2D levels and suggesting that DMP1 may regulate FGF23 expression.


Subject(s)
Bone Matrix/metabolism , Extracellular Matrix Proteins/genetics , Hypophosphatemia/genetics , Phosphates/metabolism , Phosphoproteins/genetics , Adolescent , Adult , Child , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/physiology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Homeostasis , Humans , Infant , Mutation , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Pedigree , Phosphoproteins/metabolism , Phosphoproteins/physiology
13.
Calcif Tissue Int ; 93(2): 155-62, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23700148

ABSTRACT

X-linked hypophosphatemia (XLH) is caused by mutations in the PHEX gene, which increase circulating levels of the phosphaturic hormone, fibroblast growth factor 23 (FGF23). Because XLH is a dominant disease, one mutant allele is sufficient for manifestation of the disease. However, the dosage effect of a PHEX mutation in XLH is not completely understood. To examine the effect of Phex genotypes, we compared serum biochemistries and skeletal measures between all five possible genotypes of a new murine model of XLH (Phex (K496X) or Phex (Jrt) ). Compared to sex-matched littermate controls, all Phex mutant mice had hypophosphatemia, mild hypocalcemia, and increased parathyroid hormone and alkaline phosphatase levels. Furthermore, mutant mice had markedly elevated serum Fgf23 levels due to increased Fgf23 expression and reduced cleavage of Fgf23. Although females with a homozygous Phex mutation were slightly more hypocalcemic and hypophosphatemic than heterozygous females, the two groups had comparable intact Fgf23 levels. Similarly, there was no difference in intact Fgf23 or phosphorus concentrations between hemizygous males and heterozygous females. Compared to heterozygous females, homozygous counterparts were significantly smaller and had shorter femurs with reduced bone mineral density, suggesting the existence of dosage effect in the skeletal phenotype of XLH. However, overall phenotypic trends in regards to mineral ion homeostasis were mostly unaffected by the presence of one or two mutant Phex allele(s). The lack of a gene dosage effect on circulating Fgf23 (and thus phosphorus) levels suggests that a Phex mutation may create the lower set point for extracellular phosphate concentrations.


Subject(s)
Familial Hypophosphatemic Rickets/genetics , Gene Dosage , Mutation , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Alleles , Animals , Bone Density , Female , Femur/anatomy & histology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Genotype , Heterozygote , Male , Mice , Phenotype , Phosphates/chemistry
14.
Pediatr Nephrol ; 28(4): 595-603, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23179196

ABSTRACT

Over the last decade the discovery of fibroblast growth factor 23 (FGF23) and the progressive and ongoing clarification of its role in phosphate and mineral metabolism have led to expansion of the diagnostic spectrum of primary hypophosphatemic syndromes. This article focuses on the impairment of growth in these syndromes. Growth retardation is a common, but not constant, feature and it presents with large variability. As a result of the very low prevalence of other forms of primary hypophosphatemic syndromes, the description of longitudinal growth and the pathogenesis of its impairment have been mostly studied in X-linked hypophosphatemia (XLH) patients and in Hyp mice, the animal model of this disease. In general, children with XLH have short stature with greater shortness of lower limbs than trunk. Treatment with phosphate supplements and 1α vitamin D derivatives heals active lesions of rickets, but does not normalize growth of XLH patients. Patients might benefit from recombinant human growth hormone (rhGH) therapy, which may accelerate the growth rate without increasing body disproportion or correcting hypophosphatemia. These clinical data as well as research findings obtained in Hyp mice suggest that the pathogenesis of defective growth in XLH and other hypophosphatemic syndromes is not entirely dependent on the mineralization disorder and point to other effects of hypophosphatemia itself or FGF23 on the metabolism of bone and growth plate.


Subject(s)
Body Height , Bone Development , Bone and Bones/physiopathology , Familial Hypophosphatemic Rickets/complications , Genetic Diseases, X-Linked , Growth Disorders/etiology , Phosphates/metabolism , Animals , Body Height/genetics , Bone Development/genetics , Bone and Bones/metabolism , Dietary Supplements , Disease Models, Animal , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/metabolism , Familial Hypophosphatemic Rickets/physiopathology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Growth Disorders/metabolism , Growth Disorders/physiopathology , Human Growth Hormone/therapeutic use , Humans , Mice , Mice, Transgenic , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Phosphates/therapeutic use , Recombinant Proteins/therapeutic use , Vitamins/therapeutic use
15.
Clin Calcium ; 23(10): 1463-7, 2013 Oct.
Article in Japanese | MEDLINE | ID: mdl-24076644

ABSTRACT

Bone is mineralized when hydroxyapatite crystals derived from calcium ions and inorganic phosphate (Pi) grow along collagen fibrils in the extracellular matrix. Mineralization is initiated by nucleation of those crystals. Mature osteoblasts secrete matrix vesicles into osteoid, which contain growing hydroxyapatite crystal seeds. After rupture of the lipid bilayer of those vesicles, crystals continue to grow as a mineralized nodule and adhere to collagen fibrils. It remains controversial whether nucleation occurs mainly in matrix vesicles or also extra-vesicularly around collagen fibrils. Mineralization is inhibited by pyrophosphate (PPi) and by SIBLING family proteins, which carry an acidic serine- and aspartate-rich motif (ASARM) and include osteopontin, dentin matrix protein 1 and MEPE. Intracellular and extracellular activity of these factors is regulated by the PPi-generating ectonucleotide pyrophosphatase/phosphodiesterase (ENPP1) , the PPi-transporter progressive ankylosis (ANK) protein, the PPi-degrading/Pi-generating ectoenzyme alkaline phosphatase (ALPL, TNAP) , and PHEX endopeptidase. Gain- or loss-of-function mutations in genes encoding these proteins are associated with mineralization disorders such as ectopic calcification and other pathologies.


Subject(s)
Calcification, Physiologic/physiology , Rickets/metabolism , Animals , Diphosphates/metabolism , Extracellular Matrix Proteins/metabolism , Humans , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Signal Transduction
16.
J Cell Physiol ; 227(6): 2378-87, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21826652

ABSTRACT

PHosphate-regulating gene with homology to Endopeptidase on the X chromosome (PHEX) has been identified as the gene mutated in X-linked hypophosphatemia (XLH) syndrome, the most prevalent form of rickets in humans. The predominant expression of PHEX in bones and teeth, and the defective mineralization of these tissues in XLH patients indicate that PHEX is an important regulator of mineralization. Parathyroid hormone (PTH) and PTH-related protein (PTHrP) are known to regulate the expression of numerous genes in osteoblastic cells through activation of the protein kinase A pathway, including repression of PHEX. PTH also activates the transcriptional repressor E4BP4 through the same pathway, suggesting that PTH or PTHrP-mediated repression of PHEX expression could involve E4BP4. To evaluate this possibility, we treated UMR-106 osteoblastic cells with PTHrP(1-34), and used RT-PCR and immunoblotting to analyze PHEX and E4BP4 expression. E4BP4 mRNA and protein levels were rapidly increased in cells treated with PTHrP(1-34), with a concomitant decrease in PHEX expression. This downregulation of PHEX could be reproduced by overexpression of E4BP4. Moreover, PTHrP(1-34)-mediated PHEX repression was blocked when cells were transfected with a siRNA targeting E4BP4 mRNA. Finally, DNA pull-down and luciferase assays showed that two E4BP4 response elements located in PHEX promoter were functional. These results underline the important role of E4BP4 in osteoblastic cells and further define the repression mechanism of PHEX gene by PTHrP(1-34).


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Osteoblasts/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Parathyroid Hormone-Related Protein/metabolism , Peptide Fragments/metabolism , Animals , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , Binding Sites , Blotting, Western , Down-Regulation , Genes, Reporter , Immunoprecipitation , Mice , Molecular Sequence Data , NIH 3T3 Cells , Osteoblasts/drug effects , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Phosphorylation , Promoter Regions, Genetic , Protein Binding , RNA Interference , RNA, Messenger/metabolism , Rats , Real-Time Polymerase Chain Reaction , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transfection
17.
Crit Rev Eukaryot Gene Expr ; 22(1): 61-86, 2012.
Article in English | MEDLINE | ID: mdl-22339660

ABSTRACT

More than 300 million years ago, vertebrates emerged from the vast oceans to conquer gravity and the dry land. With this transition, new adaptations occurred that included ingenious changes in reproduction, waste secretion, and bone physiology. One new innovation, the egg shell, contained an ancestral protein (ovocleidin-116) that likely first appeared with the dinosaurs and was preserved through the theropod lineage in modern birds and reptiles. Ovocleidin-116 is an avian homolog of matrix extracellular phosphoglycoprotein (MEPE) and belongs to a group of proteins called short integrin-binding ligand-interacting glycoproteins (SIBLINGs). These proteins are all localized to a defined region on chromosome 5q in mice and chromosome 4q in humans. A unifying feature of SIBLING proteins is an acidic serine aspartate-rich MEPE-associated motif (ASARM). Recent research has shown that the ASARM motif and the released ASARM peptide have regulatory roles in mineralization (bone and teeth), phosphate regulation, vascularization, soft-tissue calcification, osteoclastogenesis, mechanotransduction, and fat energy metabolism. The MEPE ASARM motif and peptide are physiological substrates for PHEX, a zinc metalloendopeptidase. Defects in PHEX are responsible for X-linked hypophosphatemic rickets (HYP). There is evidence that PHEX interacts with another ASARM motif containing SIBLING protein, dentin matrix protein-1 (DMP1). DMP1 mutations cause bone and renal defects that are identical with the defects caused by a loss of PHEX function. This results in autosomal recessive hypophosphatemic rickets (ARHR). In both HYP and ARHR, increased FGF23 expression plays a major role in the disease and in autosomal dominant hypophosphatemic rickets (ADHR), FGF23 half-life is increased by activating mutations. ASARM peptide administration in vitro and in vivo also induces increased FGF23 expression. FGF23 is a member of the fibroblast growth factor (FGF) family of cytokines, which surfaced 500 million years ago with the boney fish (i.e., teleosts) that do not contain SIBLING proteins. In terrestrial vertebrates, FGF23, like SIBLING proteins, is expressed in the osteocyte. The boney fish, however, are an-osteocytic, so a physiological bone-renal link with FGF23 and the SIBLINGs was cemented when life ventured from the oceans to the land during the Triassic period, approximately 300 million years ago. This link has been revealed by recent research that indicates a competitive displacement of a PHEX-DMP1 interaction by an ASARM peptide that leads to increased FGF23 expression. This review discusses the new discoveries that reveal a novel PHEX, DMP1, MEPE, ASARM peptide, and FGF23 bone-renal pathway. This pathway impacts not only bone formation, bone-renal mineralization, and renal phosphate homeostasis but also energy metabolism. The study of this new pathway is relevant for developing therapies for several diseases: bone-teeth mineral loss disorders, renal osteodystrophy, chronic kidney disease and bone mineralization disorders (CKD-MBD), end-stage renal diseases, ectopic arterial-calcification, cardiovascular disease renal calcification, diabetes, and obesity.


Subject(s)
Bone Diseases, Metabolic/metabolism , Bone and Bones/metabolism , Energy Metabolism/physiology , Extracellular Matrix Proteins/metabolism , Fibroblast Growth Factors/metabolism , Glycoproteins/metabolism , Kidney/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Phosphoproteins/metabolism , Animals , Bone Demineralization, Pathologic/metabolism , Bone Demineralization, Pathologic/therapy , Bone Diseases, Metabolic/therapy , Calcification, Physiologic/physiology , Chronic Kidney Disease-Mineral and Bone Disorder/metabolism , Chronic Kidney Disease-Mineral and Bone Disorder/therapy , Diabetes Mellitus/metabolism , Diabetes Mellitus/therapy , Egg Proteins/metabolism , Female , Fibroblast Growth Factor-23 , Humans , Hypophosphatemia/metabolism , Hypophosphatemia/therapy , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Male , Mechanotransduction, Cellular/physiology , Mice , Mice, Transgenic , Obesity/metabolism , Obesity/therapy , Osteocytes/metabolism , Osteomalacia/metabolism , Osteomalacia/therapy , Rickets/metabolism , Rickets/therapy
18.
FASEB J ; 25(8): 2551-62, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21507898

ABSTRACT

Fibroblastic growth factor 23 (FGF23) is a circulating phosphaturic hormone. Inactivating mutations of the endopeptidase PHEX or the SIBLING protein DMP1 result in equivalent intrinsic bone mineralization defects and increased Fgf23 expression in osteocytes. The mechanisms whereby PHEX and DMP1 regulate Fgf23 expression are unknown. We examined the possibility that PHEX and DMP1 regulate Fgf23 through a common pathway by analyzing the phenotype of compound Phex and Dmp1 mutant mice (Hyp/Dmp1(-/-)). Compared to single-mutant littermates, compound-mutant Hyp/Dmp1(-/-) mice displayed nonadditive elevations of serum FGF23 (1912 ± 183, 1715 ± 178, and 1799 ± 181 pg/ml), hypophosphatemia (P(i): 6.0 ± 0.3, 5.8 ± 0.2, and 5.4 ± 0.1 mg/dl), and severity of rickets/osteomalacia (bone mineral density: -36, -36, and -30%). Microarray analysis of long bones identified gene expression profiles implicating common activation of the FGFR pathway in all the mutant groups. Furthermore, inhibiting FGFR signaling using SU5402 in Hyp- and Dmp1(-/-)-derived bone marrow stromal cells prevented the increase in Fgf23 mRNA expression (129- and 124-fold increase in Hyp and Dmp1(-/-) vs. 1.3-fold in Hyp+SU5402 and 2.5-fold in Dmp1(-/-)+SU5402, P<0.05). For all analyses, samples collected from nonmutant wild-type littermates served as controls. These findings indicate that PHEX and DMP1 control a common pathway regulating bone mineralization and FGF23 production, the latter involving activation of the FGFR signaling in osteocytes.


Subject(s)
Extracellular Matrix Proteins/metabolism , Fibroblast Growth Factors/genetics , Osteocytes/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , Calcification, Physiologic/genetics , Calcification, Physiologic/physiology , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Familial Hypophosphatemic Rickets/etiology , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/metabolism , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Gene Expression Regulation , Kidney/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
19.
Cell Biochem Funct ; 30(5): 355-75, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22573484

ABSTRACT

The eggshell is an ancient innovation that helped the vertebrates' transition from the oceans and gain dominion over the land. Coincident with this conquest, several new eggshell and noncollagenous bone-matrix proteins (NCPs) emerged. The protein ovocleidin-116 is one of these proteins with an ancestry stretching back to the Triassic. Ovocleidin-116 is an avian homolog of Matrix Extracellular Phosphoglycoprotein (MEPE) and belongs to a group of proteins called Small Integrin-Binding Ligand Interacting Glycoproteins (SIBLINGs). The genes for these NCPs are all clustered on chromosome 5q in mice and chromosome 4q in humans. A unifying feature of the SIBLING proteins is an Acidic Serine Aspartate-Rich MEPE (ASARM)-associated motif. The ASARM motif and the released ASARM peptide play roles in mineralization, bone turnover, mechanotransduction, phosphate regulation and energy metabolism. ASARM peptides and motifs are physiological substrates for phosphate-regulating gene with homologies to endopeptidases on the X chromosome (PHEX), a Zn metalloendopeptidase. Defects in PHEX are responsible for X-linked hypophosphatemic rickets. PHEX interacts with another ASARM motif containing SIBLING protein, Dentin Matrix Protein-1 (DMP1). DMP1 mutations cause bone-renal defects that are identical with the defects caused by loss of PHEX function. This results in autosomal recessive hypophosphatemic rickets (ARHR). In both X-linked hypophosphatemic rickets and ARHR, increased fibroblast growth factor 23 (FGF23) expression occurs, and activating mutations in FGF23 cause autosomal dominant hypophosphatemic rickets (ADHR). ASARM peptide administration in vitro and in vivo also induces increased FGF23 expression. This review will discuss the evidence for a new integrative pathway involved in bone formation, bone-renal mineralization, renal phosphate homeostasis and energy metabolism in disease and health.


Subject(s)
Egg Proteins/metabolism , Fibroblast Growth Factors/metabolism , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism , Animals , Egg Proteins/chemistry , Egg Proteins/classification , Energy Metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Homeostasis , Humans , Osteogenesis , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Phosphates/metabolism
20.
Genes (Basel) ; 13(8)2022 07 28.
Article in English | MEDLINE | ID: mdl-36011266

ABSTRACT

PhexL222P mouse is a new ENU mouse model for XLH disease due to Leu to Pro amino acid modification at position 222. PhexL222P mouse is characterized by growth retardation, hypophosphatemia, hypocalcemia, reduced body bone length, and increased epiphyseal growth plate thickness and femur diameter despite the increase in PHEXL222P expression. Actually, PhexL222P mice show an increase in Fgf23, Dmp1, and Mepe and Slc34a1 (Na-Pi IIa cotransporter) mRNA expression similar to those observed in Hyp mice. Femoral osteocalcin and sclerostin and Slc34a1 do not show any significant variation in PhexL222P mice. Molecular dynamics simulations support the experimental data. P222 might locally break the E217-Q224 ß-sheet, which in turn might disrupt inter-ß-sheet interactions. We can thus expect local protein misfolding, which might be responsible for the experimentally observed PHEXL222P loss of function. This model could be a valuable addition to the existing XLH model for further comprehension of the disease occurrence and testing of new therapies.


Subject(s)
Fibroblast Growth Factors , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Animals , Bone and Bones/metabolism , Disease Models, Animal , Fibroblast Growth Factors/genetics , Mice , Mutation , PHEX Phosphate Regulating Neutral Endopeptidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL