Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
Acta Physiol (Oxf) ; 237(3): e13927, 2023 03.
Article in English | MEDLINE | ID: mdl-36606514

ABSTRACT

AIM: Perturbed calcium homeostasis limits life expectancy in familial hypomagnesaemia with hypercalciuria and nephrocalcinosis (FHHNC). This rare disease occurs by loss-of-function mutations in CLDN16 or CLDN19 genes, causing impaired paracellular reabsorption of divalent cations along the cortical thick ascending limb (cTAL). Only partial compensation takes place in the ensuing late distal convoluted tubule, connecting tubule, and collecting duct, where the luminal transient receptor potential channel V5 (TRPV5), as well as basolateral plasma membrane calcium ATPase (PMCA) and sodium-potassium exchanger (NCX1) mediate transcellular Ca2+ reabsorption. The loop diuretic furosemide induces compensatory activation in these distal segments. Normally, furosemide enhances urinary calcium excretion via inhibition of the aforementioned cTAL. As Ca2+ reabsorption in the cTAL is already severely impaired in FHHNC patients, furosemide may alleviate hypercalciuria in this disease by activation of the distal transcellular Ca2+ transport proteins. METHODS: Cldn16-deficient mice (Cldn16-/- ) served as a FHHNC model. Wild-type (WT) and Cldn16-/- mice were treated with furosemide (7 days of 40 mg/kg bw) or vehicle. We assessed renal electrolyte handling (metabolic cages) and key divalent transport proteins. RESULTS: Cldn16-/- mice show higher Ca2+ excretion than WT and compensatory stimulation of Cldn2, TRPV5, and NCX1 at baseline. Furosemide reduced hypercalciuria in Cldn16-/- mice and enhanced TRPV5 and PMCA levels in Cldn16-/- but not in WT mice. CONCLUSIONS: Furosemide significantly reduces hypercalciuria, likely via upregulation of luminal and basolateral Ca2+ transport systems in the distal nephron and collecting duct in this model for FHHNC.


Subject(s)
Furosemide , Hypercalciuria , Nephrocalcinosis , Animals , Mice , Calcium/metabolism , Carrier Proteins , Claudins/metabolism , Furosemide/pharmacology , Furosemide/therapeutic use , Hypercalciuria/drug therapy , Hypercalciuria/metabolism , Magnesium/metabolism , Nephrocalcinosis/drug therapy , Nephrocalcinosis/metabolism
2.
J Clin Endocrinol Metab ; 107(8): e3159-e3166, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35569070

ABSTRACT

BACKGROUND: Pharmacological therapy may be useful in the treatment of moderate to severe hypercalcemia in patients with infantile hypercalcemia-1 (HCINF1) due to pathogenic variants in the cytochrome P450 24 subfamily A member 1 (CYP24A1). Rifampin is an antituberculosis drug that is a potent inducer of cytochrome P450 3 subfamily A member 4, which is involved in an alternative catabolic pathway of vitamin D. The efficacy of rifampin in improving hypercalcemia was previously reported, but many questions remain on the long-term efficacy and safety. The aim of the study is to test the long-term efficacy and safety of rifampin in a patient with HCINF1. METHODS: We report clinical, biochemical, and imaging features of a 23-year-old man affected by HCINF1 with moderate hypercalcemia (12.9 mg/dL), symptomatic nephrolithiasis, nephrocalcinosis, and impaired kidney function [estimated glomerular filtration rate (eGFR) 60 mL/min/1.73 m2] treated with rifampin for an overall period of 24 months. Kidney, liver, and adrenal function were evaluated at every follow-up visit. RESULTS: In 2 months, rifampin induced a normalization of serum calcium (9.6 mg/dL) associated with an improvement of kidney function (eGFR 92 mL/min/1.73 m2) stable during the treatment. After 15 months, rifampin was temporally withdrawn because of asthenia, unrelated to impairment of adrenal function. After 3 months, the timing of drug administration was shifted from the morning to the evening, obtaining the remission of asthenia. At the end of follow-up, the nephrolithiasis disappeared and the nephrocalcinosis was stable. CONCLUSIONS: Rifampin could represent an effective choice to induce a stable reduction of calcium levels in patients with HCINF1, with a good safety profile.


Subject(s)
Hypercalcemia , Kidney Calculi , Nephrocalcinosis , Adult , Asthenia/complications , Calcium , Humans , Hypercalcemia/drug therapy , Hypercalcemia/genetics , Male , Nephrocalcinosis/drug therapy , Nephrocalcinosis/genetics , Rifampin/adverse effects , Vitamin D , Vitamin D3 24-Hydroxylase/genetics , Young Adult
3.
Front Endocrinol (Lausanne) ; 13: 800119, 2022.
Article in English | MEDLINE | ID: mdl-35250859

ABSTRACT

INTRODUCTION: Chronic hypoparathyroidism is a relatively rare disease associated with multicomponent medical therapy and various complications. The analysis of large databases of patients with chronic hypoparathyroidism is a necessary tool to enhance quality of medical care, as well as to determine the optimal clinical and therapeutic approaches, and prognostic markers of the disease. THE AIM: of this study is to estimate the clinical and biochemical profile, long-term complications, medical therapy and disease control of the patients with chronic postsurgical and non-surgical hypoparathyroidism. MATERIALS AND METHODS: the cross-sectional, observational, continuous study was based on the Russian Registry of patients with hypoparathyroidism. 544 patients from 63 regions of the Russian Federation were included in this study. RESULTS: The majority of cases had postsurgical etiology (88.4%). Postsurgical hypoparathyroidism prevailed in females (р<0.001). About a half of patients had blood calcium and phosphorus targets, 56 and 52% respectively. Nephrolithiasis was confirmed in 32.5%, nephrocalcinosis - in 12.3% of cases. The risk of nephrocalcinosis/nephrolithiasis increased by 1.85 times with disease duration more than 4.5 years. The cataract was found in 9.4%. The cut-off point for the development of cataracts was 9.5 years, with a 6.96-fold increased risk. The longer duration of hypoparathyroidism of any etiology was associated with more frequent cataract (p=0.0018).We found brain calcification in 4%, arrhythmias in 7.2% and neuropsychiatric symptoms in 5.15% of cases. Generally, the BMD in the studied group corresponded to age values, and there was no evidence for the phenomenon of high bone density. TBS was consistent with normal bone microarchitectonics. In our study, the majority of patients (83.5%) was treated with standard therapy of calcium and vitamin D supplements. 5 patients with severe disease course were treated with rhPTH (1-34). CONCLUSIONS: Analysis of the presented database indicates insufficient diagnosis of the complications associated with chronic hypoparathyroidism. Overall, hypoparathyroidism is associated with higher risks of renal stone formation, decreased GFR, cataract especially in patients with longer duration of disease.


Subject(s)
Cataract , Hypoparathyroidism , Nephrocalcinosis , Nephrolithiasis , Calcium , Cataract/complications , Cataract/drug therapy , Cross-Sectional Studies , Female , Humans , Hypoparathyroidism/complications , Hypoparathyroidism/epidemiology , Male , Nephrocalcinosis/drug therapy , Nephrolithiasis/complications , Nephrolithiasis/drug therapy , Registries
4.
Oxid Med Cell Longev ; 2022: 4345037, 2022.
Article in English | MEDLINE | ID: mdl-35251472

ABSTRACT

Intrarenal calcium oxalate (CaOx) crystals induce renal tubular epithelial cell (TEC) inflammatory and oxidative injury. This study is aimed at exploring potential therapeutic lipid components in kidney stones because lipids are involved in the development of several diseases and indicate the risk of kidney stones. Serum specimens were collected from 35 kidney stone patients and 35 normal controls. The lipid components in serum were measured, and differences were analyzed. The documented biological importance was comprehensively reviewed to identify lipids that differed significantly between the two groups to find potential agents associated with kidney stones. CaOx nephrocalcinosis mouse model was established to examine the therapeutic effects of specific lipids on CaOx deposition and CaOx-induced oxidative renal injury. Several lipids with significantly different levels were present in the serum of patients with stones and normal controls. Resolvin D1 (RvD1) (4.93-fold change, P < 0.001) and protectin D1 (PD1) (5.06-fold change, P < 0.001) were significantly decreased in the serum of patients with kidney stones, and an integrative review suggested that these factors might be associated with inflammatory responses, which is a crucial mechanism associated with stone damage. The administration of RvD1 and PD1 significantly inhibited kidney CaOx deposition and suppressed CaOx-induced renal tubular cell inflammatory injury and necrosis in a CaOx nephrocalcinosis mouse model. Furthermore, RvD1 and PD1 facilitated the expression of the oxidative indicator superoxide dismutase 2 (SOD2), inhibited NADPH oxidase 2 (NOX2) expression, and diminished intracellular reactive oxygen species (ROS) levels. This study preliminarily elucidated the role of lipids in kidney stones. The inhibitory effects of RvD1 and PD1 on oxidative damage induced by CaOx deposition provide a promising perspective for kidney stone treatment strategies.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/blood , Kidney Calculi/blood , Nephrocalcinosis/drug therapy , Signal Transduction/drug effects , Adult , Aged , Animals , Calcium Oxalate/metabolism , Case-Control Studies , Disease Models, Animal , Female , Glyoxylates/adverse effects , Humans , Kidney Tubules/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Nephrocalcinosis/chemically induced , Nephrocalcinosis/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
5.
Pediatr Nephrol ; 37(4): 907-911, 2022 04.
Article in English | MEDLINE | ID: mdl-35015123

ABSTRACT

BACKGROUND: Lumasiran, a sub-cutaneous RNA-interference therapy, has been recently approved for primary hyperoxaluria type 1 (PH1), with doses and intervals according to body weight. Little is known as to its use in infants; the aim of this study was to describe treatment outcome in 3 infants who received lumasiran therapy before 2 years of age. CASE-DIAGNOSIS/TREATMENT: Patient 1 was diagnosed antenatally and received lumasiran from day 9. According to the product information template (PIT), he received monthly lumasiran (3 times at 6 mg/kg, then 3 mg/kg), with hyperhydration and potassium citrate. Despite decreased plasma oxalate levels, persistent normal kidney function, and good tolerance, kidney ultrasound performed after 2 months found nephrocalcinosis, without normalization of urinary oxalate (UOx). The dose was increased back to 6 mg/kg, inducing a normalization in UOx. Nephrocalcinosis started to improve at month 10. Patient 2 was diagnosed at 2.5 months (acute kidney failure); nephrocalcinosis was present from diagnosis. She received monthly lumasiran (6 mg/kg), with progressive decrease in UOx and substantial improvement in kidney function but stable nephrocalcinosis after 9 injections. Patient 3 was diagnosed fortuitously (nephrocalcinosis) at 3.5 months and received lumasiran before genetic diagnosis, leading to decreased UOx and maintenance of normal kidney function. Nephrocalcinosis improved after 5 injections. CONCLUSIONS: This report presents the youngest children treated with lumasiran worldwide. Lumasiran seems effective without side effects in infants but does not completely prevent the onset of nephrocalcinosis in the most severe forms. Higher doses than those proposed in the PIT might be required because of hepatic immaturity.


Subject(s)
Hyperoxaluria, Primary , Nephrocalcinosis , Female , Humans , Hyperoxaluria, Primary/complications , Hyperoxaluria, Primary/diagnosis , Hyperoxaluria, Primary/drug therapy , Infant , Male , Nephrocalcinosis/drug therapy , Nephrocalcinosis/etiology , Oxalates , RNA, Small Interfering
6.
Osteoporos Int ; 33(6): 1385-1395, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35088103

ABSTRACT

The present study was the first prospective cohort evaluated the efficacy and safety of different doses of calcitriol in XLH children. The results suggested that a dose of 40 ng/kg/day calcitriol, compared with 20 ng/kg/day, was more effective in relieving the rickets, with similar safety outcomes. Further investigations were expected to set more dose groups. INTRODUCTION: Dose recommended for calcitriol in X-linked hypophosphatemia (XLH) varies in different studies. Therefore, we aimed to compare the efficacy as well as the safety of 20 ng/kg/d and 40 ng/kg/d calcitriol in Chinese XLH pediatrics population. METHODS: A 2-year, randomized, open-label, prospective study recruited 68 XLH children, which were randomized to receive either 40 ng/kg/day or 20 ng/kg/day calcitriol. Efficacy endpoints were the total Thacher ricket severity score (RSS) change from baseline to month 12 and 24, the difference in serum TALP level, fasting serum phosphate level, body height Z-score, and frequency of dental abscess. Safety assessments were done using renal ultrasound nephrocalcinosis grades (0-4), fasting serum and 24 h urine calcium level, and the occurrence of hyperparathyroidism. RESULTS: The decrease in the total RSS from baseline was more significant in the high-dose group at 12 (difference 0.87, p = 0.049) and 24 month (difference 1.23, p = 0.011). The serum TALP level was significantly lower in the high-dose group at 6 months. Pi level, height Z-score change, frequency of dental abscess and ratio of de novo nephrocalcinosis were comparable. A lower incidence of secondary hyperparathyroidism was seen in the high-dose group (p < 0.0001). CONCLUSION: For the first time in this prospective cohort, 40 ng/kg/d calcitriol was shown to be the more effective therapy in XLH children than the 20 ng/kg/d. Moreover, 40 ng/kg/d calcitriol was not associated with increasing adverse events. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT 03,820,518.


Subject(s)
Familial Hypophosphatemic Rickets , Hypophosphatemia , Nephrocalcinosis , Abscess/drug therapy , Calcitriol/adverse effects , Child , Familial Hypophosphatemic Rickets/drug therapy , Female , Humans , Hypophosphatemia/chemically induced , Hypophosphatemia/drug therapy , Male , Nephrocalcinosis/drug therapy , Phosphates/adverse effects , Prospective Studies
7.
Oxid Med Cell Longev ; 2021: 1676152, 2021.
Article in English | MEDLINE | ID: mdl-34512861

ABSTRACT

The roles of the lncRNA X inactive specific transcript (XIST) in many diseases, including cancers and inflammatory sickness, have been previously elucidated. However, renal calculus remained poorly understood. In this study, we revealed the potential effects of XIST on kidney stones that were exerted via inflammatory response and oxidative stress mechanisms. We established a glyoxylate-induced calcium oxalate (CaOx) stone mouse model and exposed HK-2 cells to calcium oxalate monohydrate (COM). The interactions among XIST, miR-223-3p, and NOD-like receptor protein 3 (NLRP3) and their respective effects were determined by RNAs and protein expression, luciferase activity, and immunohistochemistry (IHC) assays. Cell necrosis, reactive oxygen species (ROS) generation, and inflammatory responses were detected after silencing XIST, activating and inhibiting miR-223-3p, and both knocking down XIST and activating miR-223-3p in vitro and in vivo. The XIST, NLRP3, caspase-1, and IL-1ß levels were notably increased in kidney samples from glyoxylate-induced CaOx stone model mice. XIST knockdown significantly suppressed the inflammatory damage and ROS production and further attenuated oxalate crystal deposition. miRNA-223-3p mimics also exerted the same effects. Moreover, we verified the interactions among XIST, miRNA-223-3p and NLRP3, and the subsequent effects. Our results suggest that the lncRNA XIST participates in the formation and progression of renal calculus by interacting with miR-223-3p and the NLRP3/Caspase-1/IL-1ß pathway to mediate the inflammatory response and ROS production.


Subject(s)
MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephrocalcinosis/drug therapy , Oxidative Stress/drug effects , RNA, Long Noncoding/antagonists & inhibitors , Animals , Calcium Oxalate/administration & dosage , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Nephrocalcinosis/metabolism , Nephrocalcinosis/pathology , Transfection
8.
Endocr J ; 68(1): 111-117, 2021 Jan 28.
Article in English | MEDLINE | ID: mdl-32879144

ABSTRACT

SHORT syndrome is a rare developmental disorder frequently associated with growth failure and insulin resistant diabetes mellitus (IRDM). Since GH has a diabetogenic effect, GH therapy has been regarded as a contraindication. We observed a Brazilian girl with SHORT syndrome who received GH therapy from 4 6/12 years of age for SGA short stature. GH dosage was increased from 0.23 to 0.36 mg/kg/week, but statural response to GH therapy remained poor. Her blood HbA1c level, though it remained 5.5-6.0% in childhood, began to elevate with puberty and increased to 9.2% at 10 6/12 years of age, despite the discontinuation of GH therapy at 9 11/12 years of age. Laboratory studies indicated antibody-negative IRDM. She was treated with metformin and canagliflozin (a sodium glucose co-transporter 2 (SGLT2) inhibitor), which ameliorated overt diurnal hyperglycemia and mild nocturnal hypoglycemia and reduced her blood HbA1c around 7%. Whole exome sequencing revealed a de novo heterozygous pathogenic variant (c.1945C>T:p.(Arg649Trp)) in PIK3R1 known as the sole causative gene for SHORT syndrome. Subsequent literature review for patients with molecularly confirmed SHORT syndrome revealed the development of IRDM in 10 of 15 GH-untreated patients aged ≥12 years but in none of three GH-treated and six GH-untreated patients aged ≤10 years. These findings imply a critical role of pubertal development and/or advanced age rather than GH therapy in the development of IRDM, and a usefulness of SGLT2 inhibitor in the treatment of IRDM.


Subject(s)
Diabetes Mellitus/diagnosis , Growth Disorders/complications , Hypercalcemia/complications , Insulin Resistance/physiology , Metabolic Diseases/complications , Nephrocalcinosis/complications , Brazil , Canagliflozin/administration & dosage , Child , Diabetes Complications/diagnosis , Diabetes Complications/drug therapy , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Drug Therapy, Combination , Female , Growth Disorders/diagnosis , Growth Disorders/drug therapy , Growth Disorders/metabolism , Human Growth Hormone/administration & dosage , Humans , Hypercalcemia/diagnosis , Hypercalcemia/drug therapy , Hypercalcemia/metabolism , Metabolic Diseases/diagnosis , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Metformin/administration & dosage , Nephrocalcinosis/diagnosis , Nephrocalcinosis/drug therapy , Nephrocalcinosis/metabolism , Puberty/drug effects , Puberty/metabolism , Sodium-Glucose Transporter 2 Inhibitors/administration & dosage
9.
Anticancer Res ; 40(12): 7135-7140, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33288613

ABSTRACT

AIM: To clarify the differences in overall survival (OS) depending on the presence or absence of hypomagnesemia and the type of epidermal growth factor receptor antibody as first-line therapy for metastatic colorectal cancer (mCRC). PATIENTS AND METHODS: We retrospectively compared the OS in 68 patients who received cetuximab or panitumumab for mCRC at Ogaki Municipal Hospital (Ogaki, Japan) between January 2010 and December 2019. RESULTS: The complete and partial response rates in the cetuximab and panitumumab groups were 60.0% and 72.0%, respectively (p=0.470). The OS was significantly longer in the panitumumab group (median=1,007 days, range=208-1,433 days) than in the cetuximab group (median=735 days, range=181-2,391 days; p=0.047). Hypomagnesemia did not contribute to differences in OS in the two groups. CONCLUSION: Panitumumab may lead to a longer OS than cetuximab as first-line treatment of mCRC. The presence or absence of hypomagnesemia in cetuximab- or panitumumab-treated patients did not affect OS.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/drug therapy , ErbB Receptors/therapeutic use , Hypercalciuria/drug therapy , Nephrocalcinosis/drug therapy , Renal Tubular Transport, Inborn Errors/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/pharmacology , Colorectal Neoplasms/mortality , ErbB Receptors/pharmacology , Female , Humans , Hypercalciuria/etiology , Male , Middle Aged , Nephrocalcinosis/etiology , Renal Tubular Transport, Inborn Errors/etiology , Retrospective Studies , Survival Analysis
10.
Theranostics ; 10(16): 7319-7334, 2020.
Article in English | MEDLINE | ID: mdl-32641994

ABSTRACT

Intrarenal calcium oxalate (CaOx) crystals induce renal tubular epithelial cells (TECs) injury and inflammation, which involve Toll-like receptor 4 (TLR4)/interferon regulatory factor 1 (IRF1) signaling. Additionally, infiltrating macrophages (Mϕs) might influence intrarenal CaOx crystals and CaOx-induced renal injury. Although the roles of nuclear factor erythroid 2-related factor 2 (Nrf2) in regulating inflammation and macrophage polarization are well characterized, its potential mechanisms in regulating CaOx nephrocalcinosis remain undefined. Methods: We used a Gene Expression Omnibus dataset to analyze gene-expression profiles. Luciferase reporter, western blot, quantitative polymerase chain reaction, immunofluorescence staining, fluorescence in situ hybridization, positron emission tomography computed tomography imaging, flow cytometry, and chromatin immunoprecipitation assays were employed to study the mechanism of miR-93-TLR4/IRF1 regulation by Nrf2. Anti-inflammatory activity and regulation of macrophage polarization by Nrf2 were investigated in vitro and in vivo. Results: We found that stone-mediated kidney inflammation significantly affected stone growth, and that sulforaphane attenuated CaOx nephrocalcinosis-induced kidney injury and renal CaOx crystals deposition. Additionally, Nrf2 levels significantly increased and negatively correlated with TLR4 and IRF1 levels in a mouse model of CaOx nephrocalcinosis following sulforaphane treatment. Moreover, Nrf2 suppressed TLR4 and IRF1 levels and decreased M1-macrophage polarization which induced by supernatants from COM-stimulated TECs in vitro. In terms of mechanism, transcription factor analyses, microRNA microarray, and chromatin immunoprecipitation assays showed that Nrf2 exhibited positive transcriptional activation of miR-93-5p. In addition, Luciferase reporter, qRT-PCR, and western blot validated that miR-93-5p targets TLR4 and IRF1 mRNA. Furthermore, suppressed miR-93-5p expression partially reversed Nrf2-dependent TLR4/IRF1 downregulation. Conclusions: The results suggested that sulforaphane might promote M2Mϕ polarization and inhibit CaOx nephrocalcinosis-induced inflammatory injury to renal tubular epithelial cells via the Nrf2-miR-93-TLR4/IRF1 pathway in vitro and in vivo.


Subject(s)
Calcium Oxalate/immunology , Isothiocyanates/pharmacology , Macrophage Activation/drug effects , Nephritis/drug therapy , Nephrocalcinosis/drug therapy , Sulfoxides/pharmacology , Animals , Calcium Oxalate/chemistry , Coculture Techniques , Crystallization , Disease Models, Animal , Epithelial Cells , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Isothiocyanates/therapeutic use , Kidney Tubules/drug effects , Kidney Tubules/immunology , Kidney Tubules/pathology , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , Nephritis/immunology , Nephritis/pathology , Nephrocalcinosis/complications , Nephrocalcinosis/immunology , Primary Cell Culture , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Sulfoxides/therapeutic use , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Transcriptional Activation/immunology
11.
Mol Genet Genomic Med ; 8(9): e1385, 2020 09.
Article in English | MEDLINE | ID: mdl-32602265

ABSTRACT

BACKGROUND: SHORT syndrome is a rare inherited multisystem disease that includes characteristic facial features, growth retardation, and metabolic anomalies and is related to heterozygous mutations in the PIK3R1 gene. However, it is difficult to ascertain the relationship between the phenotype and the genotype quickly and efficiently. METHODS: We report two Chinese girls with SHORT syndrome who presented with growth retardation, dysmorphic features, insulin resistance, and diabetes. Comprehensive medical evaluations were collected, including anthropometric measurements, laboratory measurements, and imaging examinations. Whole exome and Sanger sequencing was performed to detect and confirm the underlying genetic mutations in these patients. We prescribed metformin for the patients. RESULTS: The patients both presented diabetes, insulin resistance, short stature, lipodystrophy, and characteristic facial dysmorphic features. A heterozygous mutation was detected in the PIK3R1 gene (c.1615_1617del) of Patient 1. The analysis of patient 2 revealed another PIK3R1 mutation (c.1945C>T). After family validation, neither their parents nor their brothers had similar clinical presentations or carried the same mutation. CONCLUSION: We identified two de novo heterozygous mutations in PIK3R1 as the cause of SHORT syndrome in two Chinese girls. Additionally, in terms of diabetes control, metformin works well in the early treatment stage.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/genetics , Growth Disorders/genetics , Hypercalcemia/genetics , Metabolic Diseases/genetics , Nephrocalcinosis/genetics , Adolescent , Female , Growth Disorders/drug therapy , Growth Disorders/pathology , Heterozygote , Humans , Hypercalcemia/drug therapy , Hypercalcemia/pathology , Metabolic Diseases/drug therapy , Metabolic Diseases/pathology , Metformin/therapeutic use , Mutation , Nephrocalcinosis/drug therapy , Nephrocalcinosis/pathology , Phenotype
12.
Article in English | MEDLINE | ID: mdl-32547492

ABSTRACT

Burosumab (KRN23) is an FGF23 neutralizing antibody that has been the subject of several recent clinical trials principally focused on the treatment of hypophosphatemic rickets in patients with X-linked hypophosphatemia (XLH). Since the first publications in 2014, these trials have demonstrated efficacy with minimal safety concerns in both adult and pediatric cohorts. These studies have used dose-escalation to establish a dosing regimen that is well-tolerated in clinical use. This review summarizes the clinical trial data with respect to burosumab treatment in adults and children as well as noting several clinical trials currently underway. While burosumab appears transformative for the treatment of XLH, long term follow-up studies would be required to allay concerns over the potential for nephrocalcinosis and cardiac calcification. While these do not appear to be problematic in current trials, the effects of chronic or lifelong treatment have yet to be established.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Familial Hypophosphatemic Rickets/drug therapy , Nephrocalcinosis/drug therapy , Vascular Calcification/drug therapy , Adult , Child , Clinical Trials as Topic , Familial Hypophosphatemic Rickets/pathology , Fibroblast Growth Factor-23 , Humans , Nephrocalcinosis/pathology , Vascular Calcification/pathology
13.
Am J Kidney Dis ; 73(6): 886-889, 2019 06.
Article in English | MEDLINE | ID: mdl-30765103

ABSTRACT

Mutations in the SLC34A3 gene, encoding the sodium/phosphate cotransporter 2C (NPTIIc), induce decreased renal phosphate reabsorption, hypophosphatemia, decreased fibroblast growth factor 23 and parathyroid hormone, and increased 1,25-dihydroxyvitamin D (1,25[OH]2D) levels. The complete phenotype is characterized by hypophosphatemia, hypercalciuria, and nephrolithiasis/nephrocalcinosis, leading to chronic kidney disease and osteoporosis in adults. We report a 15-year-old boy referred for nephrocalcinosis. The patient demonstrated hypercalcemia, hypercalciuria, normal serum phosphate level, normal tubular phosphate reabsorption, and increased serum 1,25(OH)2D level with suppressed serum parathyroid hormone. Compound heterozygous mutations in SLC34A3 were found. Hydrochlorothiazide failed to decrease calciuria. Fluconazole, an inhibitor of 1α-hydroxylase, was effective in normalizing calciuria without decreasing glomerular filtration rate. We conclude that children with SLC334A3 mutations can present with a less-typical phenotype, having normal serum phosphate levels and normal renal phosphate reabsorption. Genetic abnormalities of NPTIIc should be considered in cases of increased 1,25(OH)2D levels without mutations in CYP24A1. The utility of fluconazole to decrease 1,25(OH)2D levels requires confirmation in larger studies.


Subject(s)
Fluconazole/therapeutic use , Genetic Predisposition to Disease , Nephrocalcinosis/drug therapy , Nephrocalcinosis/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIc/genetics , Adolescent , Follow-Up Studies , Humans , Male , Mutation , Nephrocalcinosis/diagnosis , Pedigree , Risk Assessment , Severity of Illness Index , Treatment Outcome , Vitamin D3 24-Hydroxylase/genetics
14.
Horm Res Paediatr ; 91(4): 278-284, 2019.
Article in English | MEDLINE | ID: mdl-30227399

ABSTRACT

BACKGROUND: Under physiological conditions, proximal tubular phosphate reabsorption via NaPi-IIa (and NaPi-IIc) ensures the maintenance of phosphate homeostasis. Impairment of NaPi-IIa, encoded by SLC34A1, is associated with various overlapping clinical syndromes, including hypophosphatemic nephrolithiasis with osteoporosis, renal Fanconi's syndrome with chronic kidney disease, and idiopathic infantile hypercalcemia and nephrocalcinosis. METHODS: A patient was referred to our hospital due to hyponatremia, hyperkalemia, and hypophosphatemia, as well as persistent hypercalcemia after fluid therapy and sodium replacement. At admission to our hospital, potassium and sodium values were normal. After initiation of phosphorus therapy, hypokalemia and metabolic alkalosis were observed. Renal sonography showed bilateral medullary nephrocalcinosis. Analyses of the SLC34A1 gene were performed due to hypercalcemia and hypophosphatemia. RESULTS: Gene analyses identified a novel homozygous c.682T>C (p.W228R) (p.Trp228Arg) mutation. There are no previous reports of patients with SLC34A1 gene mutations presenting with hypokalemia and metabolic alkalosis. CONCLUSION: Herein, we present a case of infantile hypercalcemia 2 with a very different phenotype from that of previously described patients. Our findings provide further evidence for the wide range of phenotypic heterogeneity associated with NaPi-IIa impairment.


Subject(s)
Homozygote , Hypercalcemia/genetics , Mutation, Missense , Nephrocalcinosis/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Amino Acid Substitution , Female , Humans , Hypercalcemia/blood , Hypercalcemia/diagnostic imaging , Hypercalcemia/drug therapy , Infant , Nephrocalcinosis/blood , Nephrocalcinosis/diagnostic imaging , Nephrocalcinosis/drug therapy
15.
Eur J Med Genet ; 62(11): 103577, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30423445

ABSTRACT

Loss-of-function mutations in CYP24A1 (MIM 126065 20q13.2), the gene encoding the 24-hydroxylase responsible for 25-OH-D and 1,25-(OH)2D degradation, are identified in about 20% of patients presenting Idiopathic Infantile Hypercalcemia (IIH) (MIM 143880). Common features of this autosomal recessive condition included hypercalcemia with hypercalciuria, suppressed PTH and a high 25-OH-D3:24,25-(OH)2D3 ratio. Medical care mainly relies on sun protection and life-long contraindication of vitamin D to avoid complications such as early nephrocalcinosis and renal failure. Molecular diagnosis therefore keeps a crucial place in the diagnosis of IIH, and genetic counseling should be systematically recommended to prevent vitamin D administration in affected siblings. In this report is described the molecular characterization of a CYP24A1 deletion identified in two unrelated families. This highlights the potential role of CYP24A1 copy number variations (CNV) in IIH. Considering the presence of CNV affecting CYP24A1 in public databases, CNV analysis should be systematically added to the sequencing studies in IIH. Targeted Massively Parallel Sequencing (MPS) study coupled with a CNV detection tool called CovCop is a powerful method to detect genic rearrangement and improve genetic analysis.


Subject(s)
Hypercalcemia/diagnosis , Hypercalcemia/genetics , Infant, Newborn, Diseases/diagnosis , Infant, Newborn, Diseases/genetics , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/genetics , Pathology, Molecular , Vitamin D3 24-Hydroxylase/genetics , Adolescent , Child , Child, Preschool , DNA Copy Number Variations/genetics , Female , Humans , Hypercalcemia/drug therapy , Hypercalcemia/pathology , Infant , Infant, Newborn, Diseases/drug therapy , Infant, Newborn, Diseases/pathology , Loss of Function Mutation/genetics , Male , Metabolism, Inborn Errors/drug therapy , Metabolism, Inborn Errors/pathology , Nephrocalcinosis/drug therapy , Nephrocalcinosis/genetics , Nephrocalcinosis/prevention & control , Renal Insufficiency/drug therapy , Renal Insufficiency/genetics , Renal Insufficiency/prevention & control , Sequence Deletion/genetics , Vitamin D/therapeutic use
16.
Turk J Pediatr ; 60(2): 210-215, 2018.
Article in English | MEDLINE | ID: mdl-30325132

ABSTRACT

Bastug F, Nalçacioglu H, Bas VN, Tekatli-Çelik B, Çetinkaya H, Yel S. Acute renal failure due to severe hypercalcemia and nephrocalcinosis treated with two doses of pamidronate in an infant with Williams-Beuren syndrome. Turk J Pediatr 2018; 60: 210-215. Infantile hypercalcemia has been reported in 15% of infants and children with Williams-Beuren syndrome (WBS) and has generally mild clinical symptoms. However, the need for pamidronate treatment in a few infants with severe hypercalcemia associated with WBS has been reported in literature. Many disorders, such as primary hyperoxaluria, associated with nephrocalcinosis can lead to renal failure, but there are only a few reports in infants with WBS who have decreased renal function and nephrocalsinosis. We present a 23-month-old girl with WBS (confirmed with fluorescent in situ hybridization probes) who presented with acute renal failure with severe symptomatic hypercalcemia and nephrocalcinosis, which responded to two infusions of pamidronate.


Subject(s)
Acute Kidney Injury/drug therapy , Bone Density Conservation Agents/therapeutic use , Hypercalcemia/complications , Nephrocalcinosis/complications , Pamidronate/therapeutic use , Williams Syndrome/complications , Acute Kidney Injury/etiology , Female , Humans , Hypercalcemia/drug therapy , In Situ Hybridization, Fluorescence , Infant , Kidney/diagnostic imaging , Kidney/pathology , Nephrocalcinosis/drug therapy , Ultrasonography , Williams Syndrome/drug therapy
17.
Nutrition ; 38: 54-60, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28526383

ABSTRACT

OBJECTIVES: Recent studies have demonstrated that minerals play a role in glucose metabolism disorders in humans. Magnesium, in particular, is an extensively studied mineral that has been shown to function in the management of hyperglycemia, hyperinsulinemia, and insulin resistance (IR) action. The aim of this study was to investigate the effect of magnesium supplementation on IR in humans via systematic review of the available clinical trials. METHODS: This review was conducted in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) recommendations. A survey was conducted to select clinical trials related to the effects of this mineral in insulin sensitivity using the following databases: PubMed, SciVerse Scopus, ScienceDirect, and SciVerse Cochrane. RESULTS: After the selection process, 12 articles were identified as eligible, representing different clinical conditions and being free of restriction with regard to sex, age, ethnicity, and differential dosing/shape of magnesium. The results of eight clinical trials showed that supplementation with magnesium influences serum fasting glucose concentrations, and five trials determined an effect on fasting insulin levels. The results of seven studies demonstrated that mineral supplementation reduced homeostasis model assessment for IR values. CONCLUSIONS: The data of this systematic review provide evidence as to the benefits of magnesium supplementation in reducing IR in patients with hypomagnesemia presenting IR. However, new intervention studies are needed to elucidate the role of the nutrient in protection against this metabolic disorder, as well as the standardization of the type, dose, and time of magnesium supplementation.


Subject(s)
Blood Glucose/drug effects , Dietary Supplements , Hypercalciuria/drug therapy , Insulin Resistance , Magnesium/therapeutic use , Nephrocalcinosis/drug therapy , Renal Tubular Transport, Inborn Errors/drug therapy , Humans
18.
BMC Res Notes ; 10(1): 153, 2017 Apr 08.
Article in English | MEDLINE | ID: mdl-28390426

ABSTRACT

BACKGROUND: Hyperparathyroidism is common in patients undergoing kidney transplantation. Occasionally, this condition can cause early allograft dysfunction by inducing calcium phosphate deposition in the allograft, which results in nephrocalcinosis. Although nephrocalcinosis occurs occasionally in kidney allografts, it has only rarely been reported in the literature. CASE PRESENTATION: Here, we present the case of a 58-year-old Thai woman with severe hyperparathyroidism who received a living-related kidney transplant from her 35-year-old son. Our patient developed allograft dysfunction on day 2 post-transplantation despite good functioning graft on day 1. Allograft biopsy showed extensive calcium phosphate deposition in distal tubules. She was treated with cinacalcet (a calcimimetic agent) and aluminum hydroxide. Allograft function was restored to normal within 1 week after transplantation with greatly reduced intact parathyroid hormone level. CONCLUSION: Hyperparathyroidism in early functioning allograft causes elevated calcium and phosphate concentration in distal tubules resulting in nephrocalcinosis. The massive calcium phosphate precipitation obstructs tubular lumens, which leads to acute tubular dysfunction. Treatment of nephrocalcinosis with cinacalcet is safe and may improve this condition by increasing serum phosphate and reducing serum calcium and intact parathyroid hormone.


Subject(s)
Cinacalcet/therapeutic use , Hyperparathyroidism/complications , Kidney Transplantation/methods , Nephrocalcinosis/drug therapy , Adult , Allografts , Calcimimetic Agents/therapeutic use , Calcium Phosphates/metabolism , Female , Humans , Kidney Tubules, Distal/drug effects , Kidney Tubules, Distal/metabolism , Male , Middle Aged , Nephrocalcinosis/etiology , Nuclear Family , Tissue Donors , Treatment Outcome
19.
Pediatr Nephrol ; 32(7): 1123-1135, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27234911

ABSTRACT

Magnesium is essential to the proper functioning of numerous cellular processes. Magnesium ion (Mg2+) deficits, as reflected in hypomagnesemia, can cause neuromuscular irritability, seizures and cardiac arrhythmias. With normal Mg2+ intake, homeostasis is maintained primarily through the regulated reabsorption of Mg2+ by the thick ascending limb of Henle's loop and distal convoluted tubule of the kidney. Inadequate reabsorption results in renal Mg2+ wasting, as evidenced by an inappropriately high fractional Mg2+ excretion. Familial renal Mg2+ wasting is suggestive of a genetic cause, and subsequent studies in these hypomagnesemic families have revealed over a dozen genes directly or indirectly involved in Mg2+ transport. Those can be classified into four groups: hypercalciuric hypomagnesemias (encompassing mutations in CLDN16, CLDN19, CASR, CLCNKB), Gitelman-like hypomagnesemias (CLCNKB, SLC12A3, BSND, KCNJ10, FYXD2, HNF1B, PCBD1), mitochondrial hypomagnesemias (SARS2, MT-TI, Kearns-Sayre syndrome) and other hypomagnesemias (TRPM6, CNMM2, EGF, EGFR, KCNA1, FAM111A). Although identification of these genes has not yet changed treatment, which remains Mg2+ supplementation, it has contributed enormously to our understanding of Mg2+ transport and renal function. In this review, we discuss general mechanisms and symptoms of genetic causes of hypomagnesemia as well as the specific molecular mechanisms and clinical phenotypes associated with each syndrome.


Subject(s)
Arrhythmias, Cardiac/blood , Hypercalciuria/genetics , Magnesium Deficiency/genetics , Magnesium/blood , Nephrocalcinosis/genetics , Renal Elimination/genetics , Renal Reabsorption/genetics , Renal Tubular Transport, Inborn Errors/genetics , Seizures/blood , Arrhythmias, Cardiac/etiology , Child , Epithelial Sodium Channel Blockers/therapeutic use , Homeostasis/genetics , Humans , Hypercalciuria/blood , Hypercalciuria/complications , Hypercalciuria/drug therapy , Hypokalemia/blood , Hypokalemia/drug therapy , Hypokalemia/etiology , Hypokalemia/genetics , Kidney Tubules, Distal/physiology , Loop of Henle/physiology , Magnesium/physiology , Magnesium/therapeutic use , Magnesium Deficiency/complications , Magnesium Deficiency/drug therapy , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mineralocorticoid Receptor Antagonists/therapeutic use , Mitochondria/metabolism , Mutation , Nephrocalcinosis/blood , Nephrocalcinosis/complications , Nephrocalcinosis/drug therapy , Phenotype , Recommended Dietary Allowances , Renal Reabsorption/drug effects , Renal Tubular Transport, Inborn Errors/blood , Renal Tubular Transport, Inborn Errors/complications , Renal Tubular Transport, Inborn Errors/drug therapy , Seizures/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...