Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 191
Filtrar
1.
JCI Insight ; 9(9)2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38530370

RESUMO

Fibroblast growth factor 23 (FGF23) production has recently been shown to increase downstream of Gαq/11-PKC signaling in osteocytes. Inactivating mutations in the gene encoding Gα11 (GNA11) cause familial hypocalciuric hypercalcemia (FHH) due to impaired calcium-sensing receptor signaling. We explored the effect of Gα11 deficiency on FGF23 production in mice with heterozygous (Gna11+/-) or homozygous (Gna11-/-) ablation of Gna11. Both Gna11+/- and Gna11-/- mice demonstrated hypercalcemia and mildly raised parathyroid hormone levels, consistent with FHH. Strikingly, these mice also displayed increased serum levels of total and intact FGF23 and hypophosphatemia. Gna11-/- mice showed augmented Fgf23 mRNA levels in the liver and heart, but not in bone or bone marrow, and also showed evidence of systemic inflammation with elevated serum IL-1ß levels. Furin gene expression was significantly increased in the Gna11-/- liver, suggesting enhanced FGF23 cleavage despite the observed rise in circulating intact FGF23 levels. Gna11-/- mice had normal renal function and reduced serum levels of glycerol-3-phosphate, excluding kidney injury as the primary cause of elevated intact FGF23 levels. Thus, Gα11 ablation caused systemic inflammation and excess serum FGF23 in mice, suggesting that patients with FHH - at least those with GNA11 mutations - may be at risk for these complications.


Assuntos
Modelos Animais de Doenças , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Hipercalcemia , Camundongos Knockout , Animais , Feminino , Masculino , Camundongos , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Hipercalcemia/genética , Hipercalcemia/congênito , Hipercalcemia/sangue , Hipercalcemia/metabolismo , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/sangue , Fígado/metabolismo , Hormônio Paratireóideo/sangue , Hormônio Paratireóideo/metabolismo , Transdução de Sinais
2.
FEBS Open Bio ; 14(2): 290-299, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38050660

RESUMO

Congenital fibroblast growth factor 23 (FGF23)-related hypophosphatemic rickets/osteomalacia is a rare bone metabolism disorder characterized by hypophosphatemia and caused by genetic abnormalities that result in excessive secretion of FGF23. Hyp mice are a model of X-linked hypophosphatemia (XLH) caused by deletion of the PHEX gene and excessive production of FGF23. The purpose of this study was to investigate the potential of TM5614 as a therapeutic agent for the treatment of congenital FGF23-related hypophosphatemic rickets and osteomalacia in humans by administering TM5614 to Hyp mice and examining its curative effect on hypophosphatemia. After a single oral administration of TM5614 10 mg·kg-1 to female Hyp mice starting at 17 weeks of age, the serum phosphate concentration increased with a peak at 6 h after administration. ELISA confirmed that TM5614 administration decreased the intact FGF23 concentration in the blood. Expression of 25-hydroxyvitamin D-1α-hydroxylase protein encoded by Cyp27b1 mRNA in the kidney was suppressed in Hyp mice, and treatment with 10 mg·kg-1 of TM5614 normalized the expression of 25-hydroxyvitamin D-1α-hydroxylase protein and Cyp27b1 mRNA in the kidneys of these mice. Our data indicate that oral administration of TM5614 ameliorates hypophosphatemia in Hyp mice, suggesting that TM5614 may be an effective treatment for congenital FGF23-related hypophosphatemic rickets and osteomalacia.


Assuntos
Raquitismo Hipofosfatêmico Familiar , Hipofosfatemia , Osteomalacia , Camundongos , Feminino , Humanos , Animais , Raquitismo Hipofosfatêmico Familiar/tratamento farmacológico , Raquitismo Hipofosfatêmico Familiar/metabolismo , Inibidor 1 de Ativador de Plasminogênio , Osteomalacia/tratamento farmacológico , Osteomalacia/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/uso terapêutico , Hipofosfatemia/tratamento farmacológico , Hipofosfatemia/metabolismo , RNA Mensageiro/metabolismo
3.
Endocrine ; 84(1): 76-91, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38117452

RESUMO

INTRODUCTION: X-linked hypophosphatemia is an orphan disease of genetic origin and multisystem involvement. It is characterized by a mutation of the PHEX gene which results in excess FGF23 production, with abnormal renal and intestinal phosphorus metabolism, hypophosphatemia and osteomalacia secondary to chronic renal excretion of phosphate. Clinical manifestations include hypophosphatemic rickets leading to growth abnormalities and osteomalacia, myopathy, bone pain and dental abscesses. The transition of these patients to adult life continues to pose challenges to health systems, medical practitioners, patients and families. For this reason, the aim of this consensus is to provide a set of recommendations to facilitate this process and ensure adequate management and follow-up, as well as the quality of life for patients with X-linked hypophosphatemia as they transition to adult life. MATERIALS AND METHODS: Eight Latin American experts on the subject participated in the consensus and two of them were appointed as coordinators. The consensus work was done in accordance with the nominal group technique in 6 phases: (1) question standardization, (2) definition of the maximum number of choices, (3) production of individual solutions or answers, (4) individual question review, (5) analysis and synthesis of the information and (6) synchronic meetings for clarification and voting. An agreement was determined to exist with 80% votes in favor in three voting cycles. RESULTS AND DISCUSSION: Transition to adult life in patients with hypophosphatemia is a complex process that requires a comprehensive approach, taking into consideration medical interventions and associated care, but also the psychosocial components of adult life and the participation of multiple stakeholders to ensure a successful process. The consensus proposes a total of 33 recommendations based on the evidence and the knowledge and experience of the experts. The goal of the recommendations is to optimize the management of these patients during their transition to adulthood, bearing in mind the need for multidisciplinary management, as well as the most relevant medical and psychosocial factors in the region.


Assuntos
Raquitismo Hipofosfatêmico Familiar , Hipofosfatemia , Osteomalacia , Adulto , Humanos , Raquitismo Hipofosfatêmico Familiar/genética , Osteomalacia/genética , Osteomalacia/metabolismo , Consenso , Qualidade de Vida , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Fatores de Crescimento de Fibroblastos/genética
4.
Int J Rheum Dis ; 26(5): 968-972, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36625293

RESUMO

BACKGROUND: Hypophosphatemic osteomalacia (HO) is an unusual metabolic disease characterized by low concentrations of serum phosphate levels, which leads to reduced mineralization of the bone matrix. Typically, HO consists of 4 common types: X-linked dominant hypophosphatemia (XLH), autosomal dominant hypophosphatemic rickets (ADHR), tumor-induced osteomalacia (TIO), and sporadic HO. CASE PRESENTATION: We herein report the case of a 48-year-old man who developed multiple joint and bone pain and muscle weakness over 5 months with a 23-year history of psoriasis. He was diagnosed with psoriatic arthritis by primary hospitals but was unresponsive to etanercept and adalimumab treatments. After referral to our hospital, the patient was diagnosed with HOs combined with psoriasis. The patient was treated with oral phosphate solution, calcium, and active vitamin D, and the symptoms of bone and joint pain and muscle weakness gradually relieved. Since TIO accounts for the majority of adult-onset HO, positron emission tomography - computed tomography or octreotide imaging examinations had been done yearly to locate any underlying tumor in our patient, with negative findings in the 4-year follow-up. CONCLUSIONS: Diagnosis of HO remains a challenge to rheumatologists, and especially to dermatologists when accompanied by psoriasis. After excluding the inherited HO and with negative tumor, this report may be the first male case of sporadic HO combined with psoriasis.


Assuntos
Hipofosfatemia , Neoplasias , Osteomalacia , Psoríase , Adulto , Humanos , Masculino , Pessoa de Meia-Idade , Osteomalacia/etiologia , Hipofosfatemia/diagnóstico , Hipofosfatemia/etiologia , Hipofosfatemia/metabolismo , Fosfatos/metabolismo , Neoplasias/complicações , Psoríase/complicações
5.
Endocr Pract ; 29(1): 69-79, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36210014

RESUMO

OBJECTIVE: Phosphate is crucial for cell signaling, energy metabolism, nucleotide synthesis, and bone mineralization. The gut-bone-parathyroid-kidney axis is influenced by parathyroid hormone, 1,25-dihydroxyvitamin D, and phosphatonins, especially fibroblast growth factor 23 (FGF23). These hormones facilitate maintenance of phosphate homeostasis. This review summarizes current knowledge regarding the phosphate homeostasis, phosphatonin pathophysiology, and clinical implications of FGF23-related hypophosphatemic disorders, with specific focus on burosumab treatment. METHOD: A focused literature search of PubMed was conducted. RESULTS: Phosphatonins including FGF23, secreted frizzled-related protein 4, matrix extracellular phosphoglycoprotein, and fibroblast growth factor 7 play a pathogenic role in several hypophosphatemic disorders. Excess FGF23 inhibits sodium-dependent phosphate cotransporters (NaPi-2a and NaPi-2c), resulting in hyperphosphaturia and hypophosphatemia. Additionally, FGF23 suppresses 1,25-dihydroxyvitamin D synthesis in the proximal renal tubule, and thus, it indirectly inhibits intestinal phosphate absorption. Disorders of FGF23-related hypophosphatemia include X-linked hypophosphatemia (XLH), autosomal dominant hypophosphatemic rickets, autosomal recessive hypophosphatemic rickets, fibrous dysplasia/McCune-Albright syndrome, and tumor-induced osteomalacia (TIO). Complications of conventional therapy with oral phosphate and vitamin D analogs comprise gastrointestinal distress, hypercalcemia, nephrocalcinosis, and secondary/tertiary hyperparathyroidism. In both children and adults with XLH and TIO, the anti-FGF23 antibody burosumab exhibits a favorable safety profile and is associated with healing of rickets in affected children and improvement of osteomalacia in both children and adults. CONCLUSION: The treatment paradigm for XLH and TIO is changing based on data from recent clinical trials. Research suggest that burosumab is effective and safe for pediatric and adult patients with XLH or TIO.


Assuntos
Raquitismo Hipofosfatêmico Familiar , Hipofosfatemia , Osteomalacia , Adulto , Humanos , Criança , Fatores de Crescimento de Fibroblastos/metabolismo , Fosfatos/metabolismo , Fosfatos/uso terapêutico , Hipofosfatemia/tratamento farmacológico , Hipofosfatemia/etiologia , Hipofosfatemia/metabolismo , Rim/metabolismo , Osso e Ossos , Osteomalacia/tratamento farmacológico
6.
Endocr J ; 69(8): 881-896, 2022 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-35831119

RESUMO

Inorganic phosphate (Pi) in the mammalian body is balanced by its influx and efflux through the intestines, kidneys, bones, and soft tissues, at which several sodium/Pi co-transporters mediate its active transport. Pi homeostasis is achieved through the complex counter-regulatory feedback balance between fibroblast growth factor 23 (FGF23), 1,25-dihydroxyvitamin D (1,25(OH)2D), and parathyroid hormone. FGF23, which is mainly produced by osteocytes in bone, plays a central role in Pi homeostasis and exerts its effects by binding to the FGF receptor (FGFR) and αKlotho in distant target organs. In the kidneys, the main target, FGF23 promotes the excretion of Pi and suppresses the production of 1,25(OH)2D. Deficient and excess FGF23 result in hyperphosphatemia and hypophosphatemia, respectively. FGF23-related hypophosphatemic rickets/osteomalacia include tumor-induced osteomalacia and various genetic diseases, such as X-linked hypophosphatemic rickets. Coverage by the national health insurance system in Japan for the measurement of FGF23 and the approval of burosumab, an FGF23-neutralizing antibody, have had a significant impact on the diagnosis and treatment of FGF23-related hypophosphatemic rickets/osteomalacia. Some of the molecules responsible for genetic hypophosphatemic rickets/osteomalacia are highly expressed in osteocytes and function as local regulators of FGF23 production. A number of systemic factors also regulate FGF23 levels. Although the mechanisms responsible for Pi sensing in mammals have not yet been elucidated in detail, recent studies have suggested the involvement of FGFR1. The further clarification of the mechanisms by which osteocytes detect Pi levels and regulate FGF23 production will lead to the development of better strategies to treat hyperphosphatemic and hypophosphatemic conditions.


Assuntos
Raquitismo Hipofosfatêmico Familiar , Hipofosfatemia , Osteomalacia , Fosfatos , Raquitismo Hipofosfatêmico , Animais , Raquitismo Hipofosfatêmico Familiar/etiologia , Raquitismo Hipofosfatêmico Familiar/metabolismo , Fatores de Crescimento de Fibroblastos , Homeostase , Humanos , Hipofosfatemia/etiologia , Hipofosfatemia/metabolismo , Mamíferos , Osteomalacia/etiologia , Osteomalacia/metabolismo , Fosfatos/metabolismo , Raquitismo Hipofosfatêmico/etiologia , Raquitismo Hipofosfatêmico/metabolismo
7.
J Chem Inf Model ; 62(15): 3627-3637, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35868851

RESUMO

Fibroblast growth factor 23 (FGF23) is a therapeutic target for treating hereditary and acquired hypophosphatemic disorders, such as X-linked hypophosphatemic (XLH) rickets and tumor-induced osteomalacia (TIO), respectively. FGF23-induced hypophosphatemia is mediated by signaling through a ternary complex formed by FGF23, the FGF receptor (FGFR), and α-Klotho. Currently, disorders of excess FGF23 are treated with an FGF23-blocking antibody, burosumab. Small-molecule drugs that disrupt protein/protein interactions necessary for the ternary complex formation offer an alternative to disrupting FGF23 signaling. In this study, the FGF23:α-Klotho interface was targeted to identify small-molecule protein/protein interaction inhibitors since it was computationally predicted to have a large fraction of hot spots and two druggable residues on α-Klotho. We further identified Tyr433 on the KL1 domain of α-Klotho as a promising hot spot and α-Klotho as an appropriate drug-binding target at this interface. Subsequently, we performed in silico docking of ∼5.5 million compounds from the ZINC database to the interface region of α-Klotho from the ternary crystal structure. Following docking, 24 and 20 compounds were in the final list based on the lowest binding free energies to α-Klotho and the largest number of contacts with Tyr433, respectively. Five compounds were assessed experimentally by their FGF23-mediated extracellular signal-regulated kinase (ERK) activities in vitro, and two of these reduced activities significantly. Both these compounds were predicted to have favorable binding affinities to α-Klotho but not have a large number of contacts with the hot spot Tyr433. ZINC12409120 was found experimentally to disrupt FGF23:α-Klotho interaction to reduce FGF23-mediated ERK activities by 70% and have a half maximal inhibitory concentration (IC50) of 5.0 ± 0.23 µM. Molecular dynamics (MD) simulations of the ZINC12409120:α-Klotho complex starting from in silico docking poses reveal that the ligand exhibits contacts with residues on the KL1 domain, the KL1-KL2 linker, and the KL2 domain of α-Klotho simultaneously, thereby possibly disrupting the regular function of α-Klotho and impeding FGF23:α-Klotho interaction. ZINC12409120 is a candidate for lead optimization.


Assuntos
Fator de Crescimento de Fibroblastos 23 , Hipofosfatemia , Fator de Crescimento de Fibroblastos 23/antagonistas & inibidores , Humanos , Hipofosfatemia/tratamento farmacológico , Hipofosfatemia/metabolismo , Proteínas Klotho , Simulação de Acoplamento Molecular , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas
8.
Sci Rep ; 12(1): 6353, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35428804

RESUMO

Renal type II sodium-dependent inorganic phosphate (Pi) transporters NaPi2a and NaPi2c cooperate with other organs to strictly regulate the plasma Pi concentration. A high Pi load induces expression and secretion of the phosphaturic hormones parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23) that enhance urinary Pi excretion and prevent the onset of hyperphosphatemia. How FGF23 secretion from bone is increased by a high Pi load and the setpoint of the plasma Pi concentration, however, are unclear. Here, we investigated the role of Transmembrane protein 174 (Tmem174) and observed evidence for gene co-expression networks in NaPi2a and NaPi2c function. Tmem174 is localized in the renal proximal tubules and interacts with NaPi2a, but not NaPi2c. In Tmem174-knockout (KO) mice, the serum FGF23 concentration was markedly increased but increased Pi excretion and hypophosphatemia were not observed. In addition, Tmem174-KO mice exhibit reduced NaPi2a responsiveness to FGF23 and PTH administration. Furthermore, a dietary Pi load causes marked hyperphosphatemia and abnormal NaPi2a regulation in Tmem174-KO mice. Thus, Tmem174 is thought to be associated with FGF23 induction in bones and the regulation of NaPi2a to prevent an increase in the plasma Pi concentration due to a high Pi load and kidney injury.


Assuntos
Hiperfosfatemia , Hipofosfatemia , Proteínas de Membrana , Animais , Fatores de Crescimento de Fibroblastos/metabolismo , Hipofosfatemia/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Hormônio Paratireóideo , Proteínas de Transporte de Fosfato , Fosfatos/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 322(6): G561-G570, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35293263

RESUMO

Proper mitochondrial function and adequate cellular ATP are necessary for normal pancreatic protein synthesis and sorting, maintenance of intracellular organelles and enzyme secretion. Inorganic phosphate is required for generating ATP and its limited availability may lead to reduced ATP production causing impaired Ca2+ handling, defective autophagy, zymogen activation, and necrosis, which are all features of acute pancreatitis. We hypothesized that reduced dietary phosphate leads to hypophosphatemia and exacerbates pancreatitis severity of multiple causes. We observed that mice fed a low-phosphate diet before the induction of pancreatitis by either repeated caerulein administration or pancreatic duct injection as a model of pressure-induced pancreatitis developed hypophosphatemia and exhibited more severe pancreatitis than normophosphatemic mice. Pancreatitis severity was significantly reduced in mice treated with phosphate. In vitro modeling of secretagogue- and pressure-induced pancreatic injury was evaluated in isolated pancreatic acini using cholecystokinin and the mechanoreceptor Piezo1 agonist, Yoda1, under low and normal phosphate conditions. Isolated pancreatic acini were more sensitive to cholecystokinin- and Yoda1-induced acinar cell damage and mitochondrial dysfunction under low-phosphate conditions and improved following phosphate supplementation. Importantly, even mice on a normal phosphate diet exhibited less severe pancreatitis when treated with supplemental phosphate. Thus, hypophosphatemia sensitizes animals to pancreatitis and phosphate supplementation reduces pancreatitis severity. These appear to be direct effects of phosphate on acinar cells through restoration of mitochondrial function. We propose that phosphate administration may be useful in the treatment of acute pancreatitis.NEW & NOTEWORTHY Impaired ATP synthesis disrupts acinar cell homeostasis and is an early step in pancreatitis. We report that reduced phosphate availability impairs mitochondrial function and worsens pancreatic injury. Phosphate supplementation improves mitochondrial function and protects against experimental pancreatitis, raising the possibility that phosphate supplementation may be useful in treating pancreatitis.


Assuntos
Hipofosfatemia , Pancreatite , Doença Aguda , Trifosfato de Adenosina/metabolismo , Animais , Ceruletídeo/farmacologia , Colecistocinina/metabolismo , Hipofosfatemia/metabolismo , Canais Iônicos/metabolismo , Camundongos , Pâncreas/metabolismo , Pancreatite/induzido quimicamente , Pancreatite/tratamento farmacológico , Pancreatite/metabolismo , Fosfatos/metabolismo
10.
Front Endocrinol (Lausanne) ; 12: 733793, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34659120

RESUMO

Objectives: The influence of hypercortisolism on phosphate homeostasis is relatively unknown. A few previous studies have reported on patients with Cushing's syndrome (CS) with hypophosphatemia in whom serum phosphate normalized after initiation of treatment for CS. We aimed to investigate the prevalence of hypophosphatemia in CS, the association between the degree of hypercortisolism and serum phosphate and the change in serum phosphate after remission of CS. We compared the prevalence of hypophosphatemia in CS with the prevalence in the population-based Rotterdam Study (RS). Methods: Patients diagnosed with CS and treated at the Department of Endocrinology of Erasmus MC in the period of 2002-2020 were included and data was collected on age at diagnosis, sex, serum phosphate, calcium and potassium levels, kidney function and BMI. Using multivariate linear regression, we analyzed the association between 24h urinary free cortisol excretion (UFC) and serum phosphate. Changes in serum phosphate and covariates were tested with a repeated measurement ANOVA, using mean levels of laboratory values for the periods before remission, and 0-14 days and 15-180 days after remission. Results: Hypophosphatemia before treatment was present in 16% of the 99 CS patients with data on serum phosphate, 24h UFC and covariates. In comparison, the prevalence of hypophosphatemia in RS was 2.0-4.2%. Linear regression showed a negative association between the level of UFC and serum phosphate at diagnosis, which remained significant after adjusting for covariates [ß -0.002 (95%CI -0.004; -0.0004), p=0.021]. A subset of 24 patients had additional phosphate measurements at 0-14 days and 15-180 days after remission. In this subgroup, serum phosphate significantly increased from 1.03 ± 0.17 mmol/L prior to remission to 1.22 ± 0.25 mmol/L 15-180 days after remission (p = 0.008). BMI decreased after remission [-1.1 kg/m2, (95%CI -2.09 to -0.07), p=0.037]. Other covariates did not show an equivalent change over time. Conclusion: In this retrospective study, we found that 16% of patients with CS had hypophosphatemia. Moreover, serum phosphate was related to the level of cortisoluria and increased after remission of CS. Potential underlying mechanisms related to urinary phosphate excretion and possibly involving FGF23, BMI and parathyroid hormone levels should be further explored.


Assuntos
Síndrome de Cushing/epidemiologia , Hidrocortisona/metabolismo , Hipofosfatemia/epidemiologia , Fosfatos/metabolismo , Adulto , Estudos de Coortes , Síndrome de Cushing/complicações , Síndrome de Cushing/metabolismo , Síndrome de Cushing/terapia , Feminino , Homeostase/fisiologia , Humanos , Hidrocortisona/sangue , Hipofosfatemia/etiologia , Hipofosfatemia/metabolismo , Hipofosfatemia/terapia , Masculino , Pessoa de Meia-Idade , Países Baixos/epidemiologia , Fosfatos/sangue , Prevalência , Indução de Remissão , Estudos Retrospectivos
11.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34638808

RESUMO

Tissue-nonspecific alkaline phosphatase (TNAP) is mainly known for its necessary role in skeletal and dental mineralization, which relies on the hydrolysis of the mineralization inhibitor inorganic pyrophosphate (PPi). Mutations in the gene encoding TNAP leading to severe hypophosphatasia result in strongly reduced mineralization and perinatal death. Fortunately, the relatively recent development of a recombinant TNAP with a bone anchor has allowed to correct the bone defects and prolong the life of affected babies and children. Researches on TNAP must however not be slowed down, because accumulating evidence indicates that TNAP activation in individuals with metabolic syndrome (MetS) is associated with enhanced cardiovascular mortality, presumably in relation with cardiovascular calcification. On the other hand, TNAP appears to be necessary to prevent the development of steatohepatitis in mice, suggesting that TNAP plays protective roles. The aim of the present review is to highlight the known or suspected functions of TNAP in energy metabolism that may be associated with the development of MetS. The location of TNAP in liver and its function in bile excretion, lipopolysaccharide (LPS) detoxification and fatty acid transport will be presented. The expression and function of TNAP in adipocyte differentiation and thermogenesis will also be discussed. Given that TNAP is a tissue- and substrate-nonspecific phosphatase, we believe that it exerts several crucial pathophysiological functions that are just beginning to be discovered.


Assuntos
Fosfatase Alcalina/metabolismo , Metabolismo Energético , Termogênese , Adipócitos/metabolismo , Fosfatase Alcalina/genética , Animais , Bile/metabolismo , Diferenciação Celular , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Humanos , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Lipopolissacarídeos/metabolismo , Síndrome Metabólica/genética , Síndrome Metabólica/metabolismo , Camundongos , Mutação
12.
Ann Diagn Pathol ; 54: 151783, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34329884

RESUMO

BACKGROUND: Phosphaturic mesenchymal tumor-mixed connective tissue (PMT-MCT) is a rare tumor characterized clinically by presence of tumor-induced osteomalacia (TIO), subsequent to elevated fibroblastic growth factor 23 (FGF23) levels. This study aims to analyse the morphological spectrum of PMT along with clinico-pathological correlation and immunophenotype profile of this rare tumor. MATERIALS AND METHODS: Detailed histological analysis of all tumors presenting with TIO over past 7 years was done retrospectively. Immunohistochemistry was performed in all cases for SATB2, STAT6, CD34, FGF23, ERG, S100 and smooth muscle actin (SMA). RESULTS: A total of 13 cases were analysed (8 female and 5 male) with mean age of 39.8 years. Five cases were arising from bone while 4 each from soft tissue and nasal cavity/paranasal sinus. All presented with hypophosphatemia, hyperphosphaturia, elevated serum FGF23 and features suggestive of osteomalacia. Histological examination revealed basophilic 'grungy' calcification seen in 7 (53.8%), osteoid formation in 8 (61.5%), chondroid matrix in 4 (30.8%), adipose tissue in 6 (46.2%), osteoclast-like giant cells in 9 (69.2%) and hemangiopericytomatous (HPC like) blood vessels in 7 cases (53.8%). HPC like vessels and adipose tissue were more common in nasal tumors while calcification was more common in tumors arising from bone. All cases showed immunoreactivity for SATB2 and clinical improvement following resection except one case with residual tumor. CONCLUSION: PMT shows varied histological pattern with various matrix components depending on the site of the tumor. Serum FGF-23 is a useful adjunctive marker for diagnosis.


Assuntos
Mesenquimoma/metabolismo , Mesenquimoma/patologia , Osteomalacia/metabolismo , Síndromes Paraneoplásicas/metabolismo , Neoplasias de Tecidos Moles/patologia , Adulto , Biomarcadores Tumorais/análise , Feminino , Humanos , Hipofosfatemia/diagnóstico , Hipofosfatemia/metabolismo , Hipofosfatemia/patologia , Imuno-Histoquímica/métodos , Imunofenotipagem/métodos , Masculino , Mesenquimoma/diagnóstico , Pessoa de Meia-Idade , Osteomalacia/diagnóstico , Osteomalacia/patologia , Síndromes Paraneoplásicas/diagnóstico , Síndromes Paraneoplásicas/patologia , Estudos Retrospectivos , Neoplasias de Tecidos Moles/diagnóstico , Neoplasias de Tecidos Moles/metabolismo
13.
Gastroenterology ; 161(3): 982-995.e2, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34051238

RESUMO

BACKGROUND & AIMS: Heavy alcohol consumption is a common cause of acute pancreatitis; however, alcohol abuse does not always result in clinical pancreatitis. As a consequence, the factors responsible for alcohol-induced pancreatitis are not well understood. In experimental animals, it has been difficult to produce pancreatitis with alcohol. Clinically, alcohol use predisposes to hypophosphatemia, and hypophosphatemia has been observed in some patients with acute pancreatitis. Because of abundant protein synthesis, the pancreas has high metabolic demands, and reduced mitochondrial function leads to organelle dysfunction and pancreatitis. We proposed, therefore, that phosphate deficiency might limit adenosine triphosphate synthesis and thereby contribute to alcohol-induced pancreatitis. METHODS: Mice were fed a low-phosphate diet (LPD) before orogastric administration of ethanol. Direct effects of phosphate and ethanol were evaluated in vitro in isolated mouse pancreatic acini. RESULTS: LPD reduced serum phosphate levels. Intragastric administration of ethanol to animals maintained on an LPD caused severe pancreatitis that was ameliorated by phosphate repletion. In pancreatic acinar cells, low-phosphate conditions increased susceptibility to ethanol-induced cellular dysfunction through decreased bioenergetic stores, specifically affecting total cellular adenosine triphosphate and mitochondrial function. Phosphate supplementation prevented ethanol-associated cellular injury. CONCLUSIONS: Phosphate status plays a critical role in predisposition to and protection from alcohol-induced acinar cell dysfunction and the development of acute alcohol-induced pancreatitis. This finding may explain why pancreatitis develops in only some individuals with heavy alcohol use and suggests a potential novel therapeutic approach to pancreatitis. Finally, an LPD plus ethanol provides a new model for studying alcohol-associated pancreatic injury.


Assuntos
Metabolismo Energético , Hipofosfatemia/complicações , Mitocôndrias/metabolismo , Pâncreas/metabolismo , Pancreatite Alcoólica/metabolismo , Fosfatos/deficiência , Trifosfato de Adenosina/metabolismo , Animais , Modelos Animais de Doenças , Etanol , Hipofosfatemia/metabolismo , Hipofosfatemia/prevenção & controle , Masculino , Camundongos Endogâmicos C57BL , Mitocôndrias/patologia , Pâncreas/patologia , Pancreatite Alcoólica/induzido quimicamente , Pancreatite Alcoólica/patologia , Pancreatite Alcoólica/prevenção & controle , Fosfatos/administração & dosagem , Índice de Gravidade de Doença , Técnicas de Cultura de Tecidos
14.
Front Endocrinol (Lausanne) ; 12: 588096, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33716961

RESUMO

Fibroblast growth factor 23 (FGF23) has been described as an important regulator of mineral homeostasis, but has lately also been linked to iron deficiency, inflammation, and erythropoiesis. FGF23 is essential for the maintenance of phosphate homeostasis in the body and activating mutations in the gene itself or inactivating mutations in its upstream regulators can result in severe chronic hypophosphatemia, where an unbalanced mineral homeostasis often leads to rickets in children and osteomalacia in adults. FGF23 can be regulated by changes in transcriptional activity or by changes at the post-translational level. The balance between O-glycosylation and phosphorylation is an important determinant of how much active intact or inactive cleaved FGF23 will be released in the circulation. In the past years, it has become evident that iron deficiency and inflammation regulate FGF23 in a way that is not associated with its classical role in mineral metabolism. These conditions will not only result in an upregulation of FGF23 transcription, but also in increased cleavage, leaving the levels of active intact FGF23 unchanged. The exact mechanisms behind and function of this process are still unclear. However, a deeper understanding of FGF23 regulation in both the classical and non-classical way is important to develop better treatment options for diseases associated with disturbed FGF23 biology. In this review, we describe how the currently known upstream regulators of FGF23 change FGF23 transcription and affect its post-translational modifications at the molecular level.


Assuntos
Fator de Crescimento de Fibroblastos 23/genética , Fator de Crescimento de Fibroblastos 23/metabolismo , Adulto , Criança , Raquitismo Hipofosfatêmico Familiar/epidemiologia , Raquitismo Hipofosfatêmico Familiar/genética , Raquitismo Hipofosfatêmico Familiar/metabolismo , Regulação da Expressão Gênica/genética , Humanos , Hipofosfatemia/epidemiologia , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Osteomalacia/epidemiologia , Osteomalacia/genética , Osteomalacia/metabolismo , Processamento de Proteína Pós-Traducional/genética
15.
Sci Rep ; 11(1): 6175, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33731726

RESUMO

Fibroblast growth factor 23 (FGF23) is a bone-derived endocrine hormone that regulates phosphate and vitamin D metabolism. In models of FGF23 excess, renal deoxyribonuclease 1 (Dnase1) mRNA expression is downregulated. Dnase-1 is an endonuclease which binds monomeric actin. We investigated whether FGF23 suppresses renal Dnase-1 expression to facilitate endocytic retrieval of renal sodium dependent phosphate co-transporters (NaPi-IIa/c) from the brush border membrane by promoting actin polymerization. We showed that wild type mice on low phosphate diet and Fgf23-/- mice with hyperphosphatemia have increased renal Dnase1 mRNA expression while in Hyp mice with FGF23 excess and hypophosphatemia, Dnase1 mRNA expression is decreased. Administration of FGF23 in wild type and Fgf23-/- mice lowered Dnase1 expression. Taken together, our data shows that Dnase1 is regulated by FGF23. In 6-week-old Dnase1-/- mice, plasma phosphate and renal NaPi-IIa protein were significantly lower compared to wild-type mice. However, these changes were transient, normalized by 12 weeks of age and had no impact on bone morphology. Adaptation to low and high phosphate diet were similar in Dnase1-/- and Dnase1+/+ mice, and loss of Dnase1 gene expression did not rescue hyperphosphatemia in Fgf23-/- mice. We conclude that Dnase-1 does not mediate FGF23-induced inhibition of renal tubular phosphate reabsorption.


Assuntos
Desoxirribonuclease I/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Hiperfosfatemia/metabolismo , Hipofosfatemia/metabolismo , Rim/metabolismo , Fosfatos/metabolismo , Animais , Fator de Crescimento de Fibroblastos 23 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
16.
BMJ Case Rep ; 14(1)2021 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-33495177

RESUMO

Pheochromocytoma occasionally engenders catecholamine-induced hypertension crisis. Pheochromocytoma is clinically identified in 0.1%-5.7% of patients with neurofibromatosis type 1 (NF1), which is 10 times more frequently than in healthy individuals. This report describes a case of newly diagnosed NF1 presenting with pheochromocytoma crisis, with severe electrolyte depletion and deteriorating recurrent ventricular tachycardia storm. Characteristic skin lesions such as café-au-lait macules and neurofibromas contributed to the diagnosis of NF1 and pheochromocytoma. No recurrence of electrolyte depletion was found after the adrenalectomy. Primary care physicians must distinguish the characteristic skin lesions of NF1, such as café-au-lait macules and neurofibromas and recognise the risk for pheochromocytoma.


Assuntos
Neurofibromatose 1/diagnóstico , Feocromocitoma/diagnóstico , Taquicardia Ventricular/terapia , Desequilíbrio Hidroeletrolítico/terapia , 3-Iodobenzilguanidina , Adrenalectomia , Alcoolismo/complicações , Catecolaminas/urina , Cloretos/sangue , Humanos , Hipopotassemia/etiologia , Hipopotassemia/metabolismo , Hipopotassemia/terapia , Hiponatremia/etiologia , Hiponatremia/metabolismo , Hiponatremia/terapia , Hipofosfatemia/etiologia , Hipofosfatemia/metabolismo , Hipofosfatemia/terapia , Masculino , Metanefrina/urina , Pessoa de Meia-Idade , Feocromocitoma/complicações , Feocromocitoma/metabolismo , Feocromocitoma/cirurgia , Cintilografia , Compostos Radiofarmacêuticos , Taquicardia Ventricular/etiologia , Desequilíbrio Hidroeletrolítico/etiologia , Desequilíbrio Hidroeletrolítico/metabolismo
17.
Res Vet Sci ; 134: 127-136, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33360573

RESUMO

Layer fatigue syndrome caused by the lack of calcium and phosphorus can cause fracture in laying hens. The effect of phosphorus deficiency on the femur of laying hens with layer fatigue syndrome has not been studied. In this study, sixty 22-week-old Roman white layers were randomly divided into control group (group C) and low phosphorus group (group P), 30 individuals in each group. The available phosphorus content of group P was 0.18%. At the age of 26, 30 and 34 weeks, the production performance, biomechanical index, protein expression, histopathological change of femur and serological index were detected. The results showed that the laying rate, egg quality and body weight of laying hens, bone density, cortical bone thickness, rigidity, flexural modulus, flexural rigidity, the maximum load of femur and expression of osteocalcin (OCN), receptor activator of nuclear factor kappa-Β (RANK) and receptor activator of nuclear factor kappa-Β ligand (RANKL) decreased of group P. The number of osteocytes was decreased, and the voids was increased. However, cell lacunae were not obvious. The levels of phosphorus, calcium and OCN were increased, and the content of estradiol (E2), OPG and calcitonin (CT) were decreased in serum. In conclusion, the low phosphorus diet can induce layer fatigue syndrome and affect the content of OPG and E2 in serum and the expression of OCN, OPG, RANK and RANKL in femur protein, which leads to the imbalance of bone homeostasis, the thinning of femur cortex bone and the decrease of bone density.


Assuntos
Galinhas , Fêmur/patologia , Hipofosfatemia/veterinária , Doenças das Aves Domésticas/patologia , Animais , Peso Corporal , Cálcio , Dieta , Feminino , Fêmur/metabolismo , Hipofosfatemia/metabolismo , Hipofosfatemia/patologia , Fósforo/sangue , Doenças das Aves Domésticas/metabolismo
18.
BMC Nephrol ; 21(1): 339, 2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32787843

RESUMO

BACKGROUND: The nucleotide reverse transcriptase inhibitor Tenofovir Alafenamide (TAF) is a novel pro-drug of tenofovir (TFV) and possesses a superior renal safety profile compared with tenofovir disoproxil fumerate (TDF). Due to unique pharmacokinetic characteristics, treatment with TAF is not associated with significant renal proximal tubular accumulation of TFV. TAF is associated with a lower risk of acute kidney injury, chronic kidney disease, proteinuria and renal proximal tubular dysfunction than treatment with TDF. No cases of Fanconi syndrome have been reported in clinical trials of TAF. It is unknown whether treatment with TAF can lead to accumulation of TFV in proximal tubular cells and cause nephrotoxicity under certain clinical circumstances. CASE PRESENTATION: Here we report the case of a patient on stable TAF-based antiretroviral therapy with for HIV-1 infection who developed proximal tubulopathy when treated with gentamicin for febrile neutropenia in the context of relapsed Hodgkin lymphoma. Eighteen days after commencing chemotherapy for relapsed Hodgkin lymphoma the patient presented to hospital with fevers, hypotension and neutropenia. The patient was commenced on piperacillin, tazobactam and gentamicin. Within 24 h the patient developed marked hypokalaemia and hypophosphataemia requiring intravenous replacement therapy. There was proteinuria, glycosuria and evidence of marked urinary electrolyte wasting, consistent with acute proximal tubular dysfunction. Eleven days after the gentamicin was stopped the serum biochemistry normalised. The urinary electrolyte wasting and proteinuria had improved, and the glycosuria had resolved. CONCLUSION: This is the first case report to describe acute renal proximal tubulopathy in an HIV-infected patient treated with TAF and gentamicin. As the number of patients prescribed TAF outside the clinical trial setting increases, so too does the potential for previously unreported drug interactions and adverse events. Clinicians need to be aware of potential unreported adverse drug reactions as the use of TAF becomes increasingly common in clinical practice.


Assuntos
Alanina/efeitos adversos , Antibacterianos/efeitos adversos , Antivirais/efeitos adversos , Neutropenia Febril Induzida por Quimioterapia/tratamento farmacológico , Síndrome de Fanconi/induzido quimicamente , Gentamicinas/efeitos adversos , Infecções por HIV/tratamento farmacológico , Tenofovir/análogos & derivados , Doença Aguda , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neutropenia Febril Induzida por Quimioterapia/etiologia , Desprescrições , Interações Medicamentosas , Síndrome de Fanconi/metabolismo , Síndrome de Fanconi/terapia , Glicosúria/induzido quimicamente , Glicosúria/metabolismo , Glicosúria/terapia , Doença de Hodgkin/tratamento farmacológico , Humanos , Hipopotassemia/induzido quimicamente , Hipopotassemia/metabolismo , Hipopotassemia/terapia , Hipofosfatemia/induzido quimicamente , Hipofosfatemia/metabolismo , Hipofosfatemia/terapia , Masculino , Pessoa de Meia-Idade , Proteinúria/induzido quimicamente , Proteinúria/metabolismo , Proteinúria/terapia , Tenofovir/efeitos adversos
19.
Endocrinology ; 161(8)2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32591825

RESUMO

Osteomalacia is a bone-demineralizing disease of adulthood, often caused by hypovitaminosis D. Current animal models of the disease mimic osteomalacia as a consequence of gastric bypass or toxic exposure to metals, but a relevant model of diet-induced osteomalacia is lacking. For that purpose, 7-month-old female Sprague Dawley rats were randomly assigned into 2 weight-stratified groups and maintained for 4 months on synthetic diets containing negligible or normal levels of vitamin D. The dietary regimen resulted in vitamin D deficiency as measured by 25-hydroxyvitamin D serum levels; however, hypovitaminosis D per se did not affect biomarkers of calcium metabolism and bone turnover, nor did it result in increased osteoid. Thus, vitamin D depletion through the diet was found to be insufficient to induce an osteomalacia-like phenotype in the adult rat. After 4 months, the phosphate content of the vitamin D-depleted diet had decreased to 0.16% (calcium:phosphorus ratio of 5.85), resulting in an osteomalacic-like condition (trabecular osteoid surface/bone surface constituted 33%; CI, 26-40). The diet change also affected both metabolic and bone turnover biomarkers, including significantly suppressing serum fibroblast growth factor 23. Furthermore, decreased dietary phosphate in a vitamin D-depleted diet led to microarchitectural changes of trabecular and cortical bone, lower bone mass density, lower bone mass content and decreased bone strength, all indicating reduced bone quality. Taken together, our results show that osteomalacia can be induced in the adult female rat by depleting vitamin D and lowering phosphate content in the diet.


Assuntos
Hipofosfatemia/complicações , Osteomalacia/etiologia , Deficiência de Vitamina D/complicações , Animais , Remodelação Óssea , Osso e Ossos/metabolismo , Calcificação Fisiológica , Cálcio/sangue , Cálcio/urina , Feminino , Hipofosfatemia/metabolismo , Hipofosfatemia/patologia , Osteomalacia/metabolismo , Osteomalacia/patologia , Fosfatos/sangue , Fosfatos/urina , Fósforo/sangue , Fósforo/urina , Ratos , Ratos Sprague-Dawley , Vitamina D/análogos & derivados , Vitamina D/sangue , Deficiência de Vitamina D/metabolismo , Deficiência de Vitamina D/patologia
20.
Ther Apher Dial ; 24(3): 258-264, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31483921

RESUMO

The mechanism of action of fibroblast growth factor-23 (FGF23) is becoming increasingly clearer as a result of studies that have defined its structure and pleiotropic effects. Furthermore, data are emerging on the effects exerted on this hormone by iron administration. Ten main iron formulations are recognized (with clear differences in composition and possible reactions of intolerance and anaphylaxis), which are indicated for iron deficiency anemia, including nephropathic subjects, as suggested by medical guidelines. With some types of iron formulation (especially iron carboxymaltose) a particular side effect has been observed: hypophosphatemia, mediated by FGF23. This review aims to draw attention to this correlation and the contradiction represented by the presence of both positive and negative modulation by FGF23, with the effects induced by its increase even after long-term treatment with iron formulation. However, more evidence is needed to understand the reasons for this differential stimulation.


Assuntos
Anemia Ferropriva/tratamento farmacológico , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Compostos Férricos/farmacologia , Fator de Crescimento de Fibroblastos 23/metabolismo , Hipofosfatemia , Maltose/análogos & derivados , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/metabolismo , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Hematínicos/farmacologia , Humanos , Hipofosfatemia/induzido quimicamente , Hipofosfatemia/metabolismo , Hipofosfatemia/prevenção & controle , Efeitos Adversos de Longa Duração/prevenção & controle , Maltose/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...