Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Publication year range
1.
J Physiol ; 602(13): 3207-3224, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38367250

ABSTRACT

High concentrations of urinary calcium counteract vasopressin action via the activation of the Calcium-Sensing Receptor (CaSR) expressed in the luminal membrane of the collecting duct cells, which impairs the trafficking of aquaporin-2 (AQP2). In line with these findings, we provide evidence that, with respect to wild-type mice, CaSR knock-in (KI) mice mimicking autosomal dominant hypocalcaemia, display a significant decrease in the total content of AQP2 associated with significantly higher levels of AQP2 phosphorylation at Ser261, a phosphorylation site involved in AQP2 degradation. Interestingly, KI mice also had significantly higher levels of phosphorylated p38MAPK, a downstream effector of CaSR and known to phosphorylate AQP2 at Ser261. Moreover, ATF1 phosphorylated at Ser63, a transcription factor downstream of p38MAPK, was significantly higher in KI. In addition, KI mice had significantly higher levels of AQP2-targeting miRNA137 consistent with a post-transcriptional downregulation of AQP2. In vivo treatment of KI mice with the calcilytic JTT-305, a CaSR antagonist, increased AQP2 expression and reduced AQP2-targeting miRNA137 levels in KI mice. Together, these results provide direct evidence for a critical role of CaSR in impairing both short-term vasopressin response by increasing AQP2-pS261, as well as AQP2 abundance, via the p38MAPK-ATF1-miR137 pathway. KEY POINTS: Calcium-Sensing Receptor (CaSR) activating mutations are the main cause of autosomal dominant hypocalcaemia (ADH) characterized by inappropriate renal calcium excretion leading to hypocalcaemia and hypercalciuria. Current treatments of ADH patients with parathyroid hormone, although improving hypocalcaemia, do not improve hypercalciuria or nephrocalcinosis. In vivo treatment with calcilytic JTT-305/MK-5442 ameliorates most of the ADH phenotypes of the CaSR knock-in mice including hypercalciuria or nephrocalcinosis and reverses the downregulation of the vasopressin-sensitive aquaporin-2 (AQP2) expression, providing direct evidence for a critical role of CaSR in impairing vasopressin response. The beneficial effect of calcilytic in reducing the risk of renal calcification may occur in a parathyroid hormone-independent action through vasopressin-dependent inhibition of cAMP synthesis in the thick ascending limb and in the collecting duct. The amelioration of most of the abnormalities in calcium metabolism including hypercalciuria, renal calcification, and AQP2-mediated osmotic water reabsorption makes calcilytic a good candidate as a novel therapeutic agent for ADH.


Subject(s)
Aquaporin 2 , Down-Regulation , Receptors, Calcium-Sensing , Vasopressins , Animals , Aquaporin 2/metabolism , Aquaporin 2/genetics , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/genetics , Mice , Vasopressins/metabolism , Gene Knock-In Techniques , Kidney/metabolism , Kidney/drug effects , Mice, Inbred C57BL , Male , Signal Transduction , Phenotype , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/drug therapy , Calcium/metabolism , Phosphorylation , Hypocalcemia , Hypoparathyroidism/congenital
2.
Kidney Int ; 105(5): 1058-1076, 2024 May.
Article in English | MEDLINE | ID: mdl-38364990

ABSTRACT

Pathogenic variants in solute carrier family 34, member 3 (SLC34A3), the gene encoding the sodium-dependent phosphate cotransporter 2c (NPT2c), cause hereditary hypophosphatemic rickets with hypercalciuria (HHRH). Here, we report a pooled analysis of clinical and laboratory records of 304 individuals from 145 kindreds, including 20 previously unreported HHRH kindreds, in which two novel SLC34A3 pathogenic variants were identified. Compound heterozygous/homozygous carriers show above 90% penetrance for kidney and bone phenotypes. The biochemical phenotype for heterozygous carriers is intermediate with decreased serum phosphate, tubular reabsorption of phosphate (TRP (%)), fibroblast growth factor 23, and intact parathyroid hormone, but increased serum 1,25-dihydroxy vitamin D, and urine calcium excretion causing idiopathic hypercalciuria in 38%, with bone phenotypes still observed in 23% of patients. Oral phosphate supplementation is the current standard of care, which typically normalizes serum phosphate. However, although in more than half of individuals this therapy achieves correction of hypophosphatemia it fails to resolve the other outcomes. The American College of Medical Genetics and Genomics score correlated with functional analysis of frequent SLC34A3 pathogenic variants in vitro and baseline disease severity. The number of mutant alleles and baseline TRP (%) were identified as predictors for kidney and bone phenotypes, baseline TRP (%) furthermore predicted response to therapy. Certain SLC34A3/NPT2c pathogenic variants can be identified with partial responses to therapy, whereas with some overlap, others present only with kidney phenotypes and a third group present only with bone phenotypes. Thus, our report highlights important novel clinical aspects of HHRH and heterozygous carriers, raises awareness to this rare group of disorders and can be a foundation for future studies urgently needed to guide therapy of HHRH.


Subject(s)
Familial Hypophosphatemic Rickets , Hypophosphatemia , Humans , Familial Hypophosphatemic Rickets/complications , Familial Hypophosphatemic Rickets/diagnosis , Familial Hypophosphatemic Rickets/drug therapy , Hypercalciuria/diagnosis , Hypercalciuria/drug therapy , Hypercalciuria/genetics , Kidney/metabolism , Phosphates , Sodium-Phosphate Cotransporter Proteins, Type IIc/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIc/metabolism
4.
J Am Soc Nephrol ; 30(7): 1163-1173, 2019 07.
Article in English | MEDLINE | ID: mdl-31101664

ABSTRACT

BACKGROUND: The pathophysiology of genetic hypercalciuric stone-forming rats parallels that of human idiopathic hypercalciuria. In this model, all animals form calcium phosphate stones. We previously found that chlorthalidone, but not potassium citrate, decreased stone formation in these rats. METHODS: To test whether chlorthalidone and potassium citrate combined would reduce calcium phosphate stone formation more than either medication alone, four groups of rats were fed a fixed amount of a normal calcium and phosphorus diet, supplemented with potassium chloride (as control), potassium citrate, chlorthalidone (with potassium chloride to equalize potassium intake), or potassium citrate plus chlorthalidone. We measured urine every 6 weeks and assessed stone formation and bone quality at 18 weeks. RESULTS: Potassium citrate reduced urine calcium compared with controls, chlorthalidone reduced it further, and potassium citrate plus chlorthalidone reduced it even more. Chlorthalidone increased urine citrate and potassium citrate increased it even more; the combination did not increase it further. Potassium citrate, alone or with chlorthalidone, increased urine calcium phosphate supersaturation, but chlorthalidone did not. All control rats formed stones. Potassium citrate did not alter stone formation. No stones formed with chlorthalidone, and rats given potassium citrate plus chlorthalidone had some stones but fewer than controls. Rats given chlorthalidone with or without potassium citrate had higher bone mineral density and better mechanical properties than controls, whereas those given potassium citrate did not. CONCLUSIONS: In genetic hypercalciuric stone-forming rats, chlorthalidone is superior to potassium citrate alone or combined with chlorthalidone in reducing calcium phosphate stone formation and improving bone quality.


Subject(s)
Bone Density/drug effects , Calcium Phosphates/metabolism , Chlorthalidone/pharmacology , Hypercalciuria/drug therapy , Kidney Calculi/prevention & control , Potassium Citrate/pharmacology , Animals , Chlorthalidone/administration & dosage , Hypercalciuria/complications , Male , Oxalates/urine , Potassium Citrate/administration & dosage , Rats
5.
Pflugers Arch ; 471(1): 149-163, 2019 01.
Article in English | MEDLINE | ID: mdl-30109410

ABSTRACT

Hereditary hypophosphatemic rickets with hypercalciuria (HHRH; OMIM: 241530) is a rare autosomal recessive disorder with an estimated prevalence of 1:250,000 that was originally described by Tieder et al. Individuals with HHRH carry compound-heterozygous or homozygous (comp/hom) loss-of-function mutations in the sodium-phosphate co-transporter NPT2c. These mutations result in the development of urinary phosphate (Pi) wasting and hypophosphatemic rickets, bowing, and short stature, as well as appropriately elevated 1,25(OH)2D levels, which sets this fibroblast growth factor 23 (FGF23)-independent disorder apart from the more common X-linked hypophosphatemia. The elevated 1,25(OH)2D levels in turn result in hypercalciuria due to enhanced intestinal calcium absorption and reduced parathyroid hormone (PTH)-dependent calcium-reabsorption in the distal renal tubules, leading to the development of kidney stones and/or nephrocalcinosis in approximately half of the individuals with HHRH. Even heterozygous NPT2c mutations are frequently associated with isolated hypercalciuria (IH), which increases the risk of kidney stones or nephrocalcinosis threefold in affected individuals compared with the general population. Bone disease is generally absent in individuals with IH, in contrast to those with HHRH. Treatment of HHRH and IH consists of monotherapy with oral Pi supplements, while active vitamin D analogs are contraindicated, mainly because the endogenous 1,25(OH)2D levels are already elevated but also to prevent further worsening of the hypercalciuria. Long-term studies to determine whether oral Pi supplementation alone is sufficient to prevent renal calcifications and bone loss, however, are lacking. It is also unknown how therapy should be monitored, whether secondary hyperparathyroidism can develop, and whether Pi requirements decrease with age, as observed in some FGF23-dependent hypophosphatemic disorders, or whether this can lead to osteoporosis.


Subject(s)
Familial Hypophosphatemic Rickets/genetics , Hypercalciuria/genetics , Phosphates/blood , Sodium-Phosphate Cotransporter Proteins, Type IIc/genetics , Animals , Familial Hypophosphatemic Rickets/diagnosis , Familial Hypophosphatemic Rickets/drug therapy , Fibroblast Growth Factor-23 , Humans , Hypercalciuria/diagnosis , Hypercalciuria/drug therapy , Loss of Function Mutation , Phosphates/therapeutic use , Sodium-Phosphate Cotransporter Proteins, Type IIc/metabolism , Vitamin D/blood , Vitamin D/therapeutic use , Vitamins/blood , Vitamins/therapeutic use
6.
Dev Period Med ; 22(2): 145-152, 2018.
Article in English | MEDLINE | ID: mdl-30056401

ABSTRACT

AIM: To study the impact of vitamin D supplementation on vitamin D concentration in plasma, calcium urinary excretion and bone density in patients with urolithiasis in the course of idiopathic hypercalciuria and with a low vitamin D level. MATERIALS AND METHOD: Prospective analysis concerning 28 patients (16 boys, 12 girls) aged 6-14 years (average 10.4) in terms of urinary calcium excretion (mg/kg/day and Ca/Creatinine ratio in morning urine sample), 25OHD blood level after 3, 6, 9 and 12 months of individually recommended doses of vitamin D supplementation (400 IU or 800 IU). The doses were determined on the basis of 25 (OH) D deficiency. The children were on a normocalcemic diet. The bone mineral density of the patients was assessed before and after 12 months of vitamin D use at the aforementioned doses. RESULTS: There was no statistically significant correlation between 25 (OH) D plasma concentration and urinary calcium excretion measured on Ca /Creatinine ratio in daily urine collection and Ca/Creatinine ratio in the morning urine sample. No statistically significant change in calcium excretion was noted (measured by calciuria in daily urine collection and the calcium to creatinine ratio in the morning urine sample). A statistically significant increase in vitamin D plasma concentration was observed. Improvement in bone mineral density was not statistically significant. CONCLUSIONS: Supplementation of vitamin D in the children with idiopathic hypercalciuria and urolithiasis who were examined seems to be safe. The decision to start treatment and the selection of the vitamin D dose should be considered individually. Patients with urolithiasis should be carefully monitored for calcium/phosphate metabolism parameters and the activity of the disease. Supplementation of low doses vitamin D in the children examined did not improve bone mineral density.


Subject(s)
Bone Density/drug effects , Calcium, Dietary/urine , Hypercalciuria/drug therapy , Vitamin D Deficiency/drug therapy , Vitamin D/blood , Vitamin D/pharmacology , Adolescent , Child , Dietary Supplements , Female , Humans , Male , Prospective Studies , Urolithiasis , Vitamin D/therapeutic use
7.
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
8.
Urologiia ; (6): 112-119, 2017 Dec.
Article in Russian | MEDLINE | ID: mdl-29376607

ABSTRACT

Most patients with idiopathic hypercalciuria and calcium nephrolithiasis have a family history of the disease. Idiopathic hypercalciuria is a metabolic abnormality with various causes and developmental pathways. The systematic review describes specific mutations associated with idiopathic hypercalciuria and nephrolithiasis. Detection of these mutations may provide a better understanding of the pathogenesis of this heterogeneous disease and personalize patient management depending on the detected polymorphisms. A promising treatment option for a mutation in the vitamin D receptor gene is thiazide diuretics in combination with bisphosphonates. Among bisphosphonates, the drug of choice which has been most strongly supported by research evidence is alendronate.


Subject(s)
Alendronate/therapeutic use , Mutation , Nephrolithiasis , Receptors, Calcitriol , Female , Humans , Hypercalciuria/diagnosis , Hypercalciuria/drug therapy , Hypercalciuria/genetics , Hypercalciuria/metabolism , Male , Nephrolithiasis/diagnosis , Nephrolithiasis/drug therapy , Nephrolithiasis/genetics , Nephrolithiasis/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism
9.
J Pharmacol Sci ; 130(4): 204-11, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26980429

ABSTRACT

Notwithstanding compelling contribution of NF-κB to the progression of osteoporosis has been reported, little is known regarding direct inhibition of NF-κB benefiting osteoporosis. In this study, therefore, we evaluated the role of celastrol, an NF-κB inhibitor, in a mouse model of secondary osteoporosis. Animals were divided into three groups as Sham (control), SO (secondary osteoporosis) and SO + CA (secondary osteoporosis treated with celastrol). Significant decreases in body weight and body fat were observed following celastrol treatment in SO group, but leptin levels were much higher. Celastrol also exhibited a significant decrease in urinary calcium excretion. Moreover, other important events were observed after celastrol treatment, covering substantial decrements in serum concentrations of PTH, TRAP-5b, CTX and DPD, improved structure of articular cartilage and cancellous bone (revealed by H&E and safranin-O staining), and significant decline in levels of NF-κB (P65), MMP-1, and MMP-9. These findings demonstrated that celastrol treatment not only improved abnormal lipid metabolism and hypercalciuria in mice subjected to secondary osteoporosis, but also ameliorated articular cartilage lesions. Our results provided evidence of targeted therapy for NF-κB in the clinical treatment of secondary osteoporosis.


Subject(s)
Cartilage, Articular , Hypercalciuria/drug therapy , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Osteoporosis/drug therapy , Osteoporosis/genetics , Triterpenes/pharmacology , Adipose Tissue/metabolism , Animals , Body Weight/drug effects , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Disease Models, Animal , Disease Progression , Leptin/metabolism , Lipid Metabolism/drug effects , Male , Mice, Inbred C57BL , Molecular Targeted Therapy , Parathyroid Hormone/blood , Pentacyclic Triterpenes , Triterpenes/therapeutic use
11.
Curr Opin Nephrol Hypertens ; 24(4): 336-44, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26050120

ABSTRACT

PURPOSE OF REVIEW: In this review, we discuss how the genetic hypercalciuric stone-forming (GHS) rats, which closely model idiopathic hypercalciuria and stone formation in humans, provide insights into the pathophysiology and consequences of clinical hypercalciuria. RECENT FINDINGS: Hypercalciuria in the GHS rats is due to a systemic dysregulation of calcium transport, as manifest by increased intestinal calcium absorption, increased bone resorption and decreased renal tubule calcium reabsorption. Increased levels of vitamin D receptor in intestine, bone and kidney appear to mediate these changes. The excess receptors are biologically active and increase tissue sensitivity to exogenous vitamin D. Bones of GHS rats have decreased bone mineral density (BMD) as compared with Sprague-Dawley rats, and exogenous 1,25(OH)2D3 exacerbates the loss of BMD. Thiazide diuretics improve the BMD in GHS rats. SUMMARY: Studying GHS rats allows direct investigation of the effects of alterations in diet and utilization of pharmacologic therapy on hypercalciuria, urine supersaturation, stone formation and bone quality in ways that are not possible in humans.


Subject(s)
Calcium, Dietary/metabolism , Hypercalcemia/metabolism , Hypercalciuria/metabolism , Kidney Calculi/metabolism , Receptors, Calcitriol/metabolism , Animals , Disease Models, Animal , Humans , Hypercalcemia/drug therapy , Hypercalcemia/genetics , Hypercalciuria/drug therapy , Kidney Calculi/drug therapy , Kidney Calculi/genetics
12.
Am J Physiol Renal Physiol ; 306(9): F1081-7, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24573387

ABSTRACT

Genetic hypercalciuric stone-forming (GHS) rats demonstrate increased intestinal Ca absorption, increased bone resorption, and reduced renal tubular Ca reabsorption leading to hypercalciuria and all form kidney stones. GHS have increased vitamin D receptors (VDR) at these sites of Ca transport. Injection of 1,25(OH)2D3 (1,25D) leads to a greater increase in urine (u)Ca in GHS than in control Sprague-Dawley (SD), possibly due to the additional VDR. In GHS the increased uCa persists on a low-Ca diet (LCD) suggesting enhanced bone resorption. We tested the hypothesis that LCD, coupled to inhibition of bone resorption by alendronate (alen), would eliminate the enhanced 1,25D-induced hypercalciuria in GHS. SD and GHS were fed LCD and half were injected daily with 1,25D. After 8 days all were also given alen until euthanasia at day 16. At 8 days, 1,25D increased uCa in SD and to a greater extent in GHS. At 16 days, alen eliminated the 1,25D-induced increase in uCa in SD. However, in GHS alen decreased, but did not eliminate, the 1,25D-induced hypercalciuria, suggesting maximal alen cannot completely prevent the 1,25D-induced bone resorption in GHS, perhaps due to increased VDR. There was no consistent effect on mRNA expression of renal transcellular or paracellular Ca transporters. Urine CaP and CaOx supersaturation (SS) increased with 1,25D alone in both SD and GHS. Alen eliminated the increase in CaP SS in SD but not in GHS. If these results are confirmed in humans with IH, the use of bisphosphonates, such as alen, may not prevent the decreased bone density observed in these patients.


Subject(s)
Alendronate/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Resorption/prevention & control , Calcitriol , Calcium, Dietary/urine , Hypercalciuria/drug therapy , Kidney Calculi/drug therapy , Kidney/metabolism , Animals , Bone Density/drug effects , Bone Resorption/chemically induced , Bone Resorption/genetics , Bone Resorption/urine , Calcium, Dietary/administration & dosage , Disease Models, Animal , Genotype , Hypercalciuria/chemically induced , Hypercalciuria/genetics , Hypercalciuria/urine , Intestinal Absorption , Intestinal Mucosa/metabolism , Kidney Calculi/chemically induced , Kidney Calculi/genetics , Kidney Calculi/urine , Male , Phenotype , Rats , Rats, Sprague-Dawley , Time Factors
13.
Curr Opin Urol ; 24(6): 633-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25188231

ABSTRACT

PURPOSE OF REVIEW: To establish the relationship between calcium nephrolithiasis, bone densitometry scoring, and bone mineral density (BMD) loss according to bone turnover markers (BTMs) and urinary metabolites. RECENT FINDINGS: Patients with recurrent calcium nephrolithiasis and idiopathic fasting hypercalciuria (urinary calcium/creatinine ratio >0.11) are more likely to have BMD loss that may lead to osteopenia or osteoporosis. In these patients, BTMs may be used as a surrogate for both bone health and stone recurrence. Suspect higher lithogenic states when calcium stone formers have serum beta-crosslaps (resorptive marker) greater than 0.311 ng/ml, serum osteocalcin (formative marker) greater than 13.2 ng/ml, and beta-crosslaps/osteocalcin ratio greater than 0.024. SUMMARY: Patients with recurrent calcium nephrolithiasis and fasting hypercalciuria have a higher incidence of osteopenia and osteoporosis, measured by the dual-energy X-ray absorptiometry. These patients present not only with hypercalciuria and increased BTMs (mainly resorptive), but also up to 30% have hypocitraturia and increased urinary calcium/citrate ratio (>0.25). On the basis of these results, a diagnostic algorithm was created, classifying hypercalciurics according to their fasting calcium/creatinine and calcium/citrate ratio. Medical therapy for these patients is aimed at improving the dietary habits (normocalcemic, low salt, low animal protein diet), prescribing combinations of potassium citrate, thiazides, and bisphosphonates, and correcting bone and urinary abnormalities that may lower future skeletal and kidney stone risk.


Subject(s)
Bone Demineralization, Pathologic/metabolism , Calcium/metabolism , Hypercalciuria/metabolism , Nephrolithiasis/metabolism , Osteoporosis/metabolism , Bone Demineralization, Pathologic/diagnosis , Bone Demineralization, Pathologic/drug therapy , Humans , Hypercalciuria/diagnosis , Hypercalciuria/drug therapy , Nephrolithiasis/diagnosis , Nephrolithiasis/drug therapy , Osteoporosis/diagnosis , Osteoporosis/drug therapy
14.
BMC Endocr Disord ; 14: 81, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25292184

ABSTRACT

BACKGROUND: Familial Hyperparathyroidism (HPT) and Familial benign Hypocalciuric Hypercalcemia (FHH) are the most common causes of hereditary hypercalcemia. FHH has been demonstrated to be caused by inactivating mutations of calcium-sensing receptor (CaSR) gene, involved in PTH regulation as well as in renal calcium excretion. CASE PRESENTATION: In two individuals, father and son, we found a novel heterozygous mutation in CaSR gene. The hypercalcemia was present only in father, which, by contrast to the classic form of FHH showed hypercalciuria (from 300 to 600 mg/24 h in different evaluations) and a Calcium/Creatinine ratio of 0.031, instead of low or normal calciuria (<0.01 typical finding in FHH). His son showed the same mutation in CaSR gene, but no clinical signs or hypercalcemia although serum ionized calcium levels were close to the upper limit of normal values (1.30 mmol/L: normal range: 1.12-1.31 mmol/L). Sequence analysis revealed a point mutation at codon 972 of CaSR gene (chromosome 3q), located within cytoplasmic domain of the CaSR, that changes Threonine with Methionine. The father was treated with Cinacalcet 90 mg/day, with a decrease of total serum calcemia from an average value of 12.2 mg/dl to 10.9 mg/dl. CONCLUSION: This is a case of a novel inactivating point mutation of CaSR gene that determines an atypical clinical presentation of FHH, characterized by hypercalcemia, hypercalciuria and inadequate normal PTH levels. Functional assay demonstrated that the 972 M variant influenced the maturation of the protein, in terms of the post-translational glycosylation. The impairment of the receptor activity is in keeping with the specific localization of the 972 residue in the C-terminal tail, assigned to the intracellular signalling, that on the basis of the our findings appears to be differently modulated in parathyroid gland and in kidney.


Subject(s)
Calcimimetic Agents/therapeutic use , Hypercalcemia/congenital , Hypercalcemia/genetics , Hypercalciuria/genetics , Naphthalenes/therapeutic use , Parathyroid Hormone/genetics , Point Mutation , Receptors, Calcium-Sensing/genetics , Adult , Aged , Blotting, Western , Cinacalcet , Genetic Markers/genetics , Humans , Hypercalcemia/diagnosis , Hypercalcemia/drug therapy , Hypercalciuria/diagnosis , Hypercalciuria/drug therapy , Male , Parathyroid Hormone/metabolism , Pedigree , Receptors, Calcium-Sensing/metabolism , Treatment Outcome
15.
J Pediatr Hematol Oncol ; 36(4): 305-7, 2014 May.
Article in English | MEDLINE | ID: mdl-23426003

ABSTRACT

Cetuximab, a monoclonal antibody specific for epidermal growth factor receptor, is increasingly used off-label and in early-phase trials for pediatric malignancies. Here, we report a patient with metastatic medulloblastoma receiving therapy with cyclophosphamide, vinblastine, and cetuximab. During evaluation for possible seizures, he was noted to be severely hypocalcemic, hypokalemic, and hypomagnesemic, a consequence of the blockade of renal epidermal growth factor receptor expression. His symptoms rapidly abated with intravenous electrolyte repletion. This case highlights the clinical heterogeneity of tetany and the importance of careful laboratory screening for known adverse effects of chemotherapy, particularly when newer biological agents are used off-study in combination chemotherapeutic regimens.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Cerebellar Neoplasms/drug therapy , Electrolytes/administration & dosage , Hypercalciuria , Medulloblastoma/drug therapy , Nephrocalcinosis , Renal Tubular Transport, Inborn Errors , Tetany/drug therapy , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Cerebellar Neoplasms/pathology , Cetuximab , Child, Preschool , Humans , Hypercalciuria/chemically induced , Hypercalciuria/drug therapy , Male , Medulloblastoma/pathology , Neoplasm Metastasis , Nephrocalcinosis/chemically induced , Nephrocalcinosis/drug therapy , Renal Tubular Transport, Inborn Errors/chemically induced , Renal Tubular Transport, Inborn Errors/drug therapy , Seizures/chemically induced , Seizures/drug therapy , Tetany/chemically induced
16.
Acta Paediatr ; 103(11): 1117-25, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25039540

ABSTRACT

UNLABELLED: Autosomal dominant hypocalcaemia (ADH) is caused by activating variants in the calcium-sensing receptor (CASR) gene, but detailed information on the paediatric phenotype is limited. The current paper presents a case of severe ADH and systematically reviews the literature on ADH in children. CONCLUSION: We found that the severity of clinical neurological symptoms was inversely related to serum calcium levels and a high prevalence of renal calcifications and/or basal ganglia calcifications in children with ADH.


Subject(s)
Hypercalciuria/genetics , Hypocalcemia/genetics , Hypoparathyroidism/congenital , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/physiology , Calcium/therapeutic use , Female , Genetic Variation , Humans , Hypercalciuria/diagnosis , Hypercalciuria/drug therapy , Hypocalcemia/diagnosis , Hypocalcemia/drug therapy , Hypoparathyroidism/diagnosis , Hypoparathyroidism/drug therapy , Hypoparathyroidism/genetics , Infant
17.
Am J Physiol Renal Physiol ; 305(4): F592-9, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23720347

ABSTRACT

The most common metabolic abnormality found in calcium (Ca) kidney stone formers is idiopathic hypercalciuria (IH). Using endogenous lithium (Li) clearance, we previously showed that in IH, there is decreased proximal tubule sodium absorption, and increased delivery of Ca into the distal nephron. Distal Ca reabsorption may facilitate the formation of Randall's plaque (RP) by washdown of excess Ca through the vasa recta toward the papillary tip. Elevated Ca excretion leads to increased urinary supersaturation (SS) with respect to calcium oxalate (CaOx) and calcium phosphate (CaP), providing the driving force for stone growth on RP. Thiazide (TZ) diuretics reduce Ca excretion and prevent stone recurrence, but the mechanism in humans is unknown. We studied the effect of chronic TZ administration on renal mineral handling in four male IH patients using a fixed three meal day in the General Clinical Research Center. Each subject was studied twice: once before treatment and once after 4-7 mo of daily chlorthalidone treatment. As expected, urine Ca fell with TZ, along with fraction of filtered Ca excreted. Fraction of filtered Li excreted also fell sharply with TZ, as did distal delivery of Ca. Unexpectedly, TZ lowered urine pH. Together with reduced urine Ca, this led to a marked fall in CaP SS, but not CaOx SS. Since CaOx stone formation begins with an initial CaP overlay on RP, by lowering urine pH and decreasing distal nephron Ca delivery, TZ might diminish stone risk both by reducing CaP SS, as well as slowing progression of RP.


Subject(s)
Chlorthalidone/administration & dosage , Diuretics/administration & dosage , Hypercalciuria/drug therapy , Kidney Tubules, Proximal/drug effects , Thiazides/administration & dosage , Adult , Calcium/blood , Calcium/urine , Humans , Hypercalciuria/metabolism , Kidney Tubules, Proximal/metabolism , Male , Middle Aged
18.
J Pediatr Endocrinol Metab ; 36(5): 500-504, 2023 May 25.
Article in English | MEDLINE | ID: mdl-36935580

ABSTRACT

OBJECTIVES: Autosomal dominant hypocalcaemia 1 (ADH1) is a rare autosomal dominant genetic disease, due to the activating mutations of the calcium-sensing receptor (CASR) gene. The current paper presents a severe case of ADH1 with intellectual backwardness, and systematically reviews the reported 17 ADH1 patients in China. CASE PRESENTATION: A 7 years old boy with recurrent seizures over 1 year was admitted at Yuying children' hospital, the clinical centre of south province of Zhejiang. Auxiliary examinations demonstrated hypocalcaemia, hyperphosphatemia, hypomagnesemia, hypercalciuria, low parathyroid hormone (PTH), basal ganglia calcifications, normal range of serum creatinine, and 25-hydroxyvitamin D. Wechsler's intelligence test result indicated intellectually backward. The patient's genotype found a heterozygous variant in CASR gene, c.T416C p. (Ile139Thr). This article also systematically reviews the literatures on ADH1 in China and summarises the clinical characteristics and treatment. CONCLUSIONS: ADH1 can be a cause of idiopathic hypoparathyroidism. Recognition and rational treatment is important for symptom improvement and reducing high potential adverse effects.


Subject(s)
Hypocalcemia , Hypoparathyroidism , Male , Child , Humans , Hypocalcemia/genetics , Receptors, Calcium-Sensing/genetics , Hypercalciuria/drug therapy , Hypercalciuria/genetics , China , Mutation , Calcium
19.
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
20.
J Nephrol ; 36(1): 69-82, 2023 01.
Article in English | MEDLINE | ID: mdl-35696042

ABSTRACT

BACKGROUND: Renal sarcoidosis is a rare cause of tubulointerstitial nephritits (TIN). The clinical and pathological characteristics, as well as outcomes, of renal sarcoidosis remain unclear. METHODS: This single-center study retrospectively analyzed 18 patients affected by sarcoidosis with tubulointerstitial nephritis (TIN) and 53 patients with tubulointerstitial nephritis  not related to sarcoidosis. Patients were further stratified into the granulomatous (12 sarcoidosis and 6 non-sarcoidosis) and non-granulomatous (6 sarcoidosis and 47 non-sarcoidosis) TIN groups. RESULTS: Half of the patients with renal sarcoidosis had signs of acute kidney injury at kidney biopsy, 94% of whom presented with extra-renal involvement. The prevalence of hypercalcemia, hypercalciuria, and elevated serum angiotensin-converting enzyme levels was 27.6%, 33.3%, and 31.3%, respectively. Renal sarcoidosis patients with eGFR < 30 mL/min/1.73 m2 scored higher for total chronic tubulointerstitial injury (p = 0.044) and glomerular sclerosis (p = 0.027). Compared to non-sarcoidosis patients, higher urinary calcium levels (for patients with GFR [Formula: see text] 40 mL/min/1.73 m2, p = 0.034), lower scores of acute tubular injury (p = 0.008), and more prominent glomerular sclerosis were observed in renal sarcoidosis. Similar characteristics of chronicity and hypercalciuria were also identified in granulomatous interstitial nephritis; however, interstitial inflammation was obvious (p = 0.001). Patients with renal sarcoidosis were initially treated with corticosteroids. Five patients receiving immunosuppressive agents showed better long-term renal recovery. High 24-h urine calcium (adjusted by weight) was identified as a factor associated with long-term remission. CONCLUSION: Renal sarcoidosis is a systemic disease of insidious onset and chronic progression, sharing similar features of chronicity and hypercalciuria with granulomatous interstitial nephritis of other cause. Hypercalciuria may predict a better response to immunosuppressive therapy, presumably indicating active interstitial inflammation; thus, strengthened immunosuppression might be considered.


Subject(s)
Hypercalcemia , Nephritis, Interstitial , Sarcoidosis , Humans , Calcium , Hypercalcemia/etiology , Hypercalcemia/complications , Hypercalciuria/drug therapy , Hypercalciuria/complications , Immunosuppression Therapy , Inflammation/complications , Kidney/pathology , Nephritis, Interstitial/diagnosis , Nephritis, Interstitial/drug therapy , Retrospective Studies , Sarcoidosis/complications , Sarcoidosis/drug therapy , Sarcoidosis/diagnosis , Sclerosis/complications , Sclerosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL