Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Publication year range
1.
Curr Opin Nephrol Hypertens ; 32(4): 359-365, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37074688

ABSTRACT

PURPOSE OF REVIEW: Most kidney stones are composed of calcium, and the greatest risk factor for kidney stone formation is hypercalciuria. Patients who form kidney stones often have reduced calcium reabsorption from the proximal tubule, and increasing this reabsorption is a goal of some dietary and pharmacological treatment strategies to prevent kidney stone recurrence. However, until recently, little was known about the molecular mechanism that mediates calcium reabsorption from the proximal tubule. This review summarizes newly uncovered key insights and discusses how they may inform the treatment of kidney stone formers. RECENT FINDINGS: Studies examining claudin-2 and claudin-12 single and double knockout mice, combined with cell culture models, support complementary independent roles for these tight junction proteins in contributing paracellular calcium permeability to the proximal tubule. Moreover, a family with a coding variation in claudin-2 causing hypercalciuria and kidney stones have been reported, and reanalysis of Genome Wide Association Study (GWAS) data demonstrates an association between noncoding variations in CLDN2 and kidney stone formation. SUMMARY: The current work begins to delineate the molecular mechanisms whereby calcium is reabsorbed from the proximal tubule and suggests a role for altered claudin-2 mediated calcium reabsorption in the pathogenesis of hypercalciuria and kidney stone formation.


Subject(s)
Calcium , Hypercalciuria , Kidney Calculi , Kidney Calculi/genetics , Kidney Calculi/physiopathology , Kidney Calculi/prevention & control , Kidney Calculi/therapy , Hypercalciuria/genetics , Hypercalciuria/physiopathology , Hypercalciuria/prevention & control , Hypercalciuria/therapy , Calcium/metabolism , Humans , Animals , Claudin-2/genetics , Claudin-2/metabolism , Claudins/genetics , Claudins/metabolism , Genome-Wide Association Study , Kidney Tubules, Proximal/physiopathology
2.
BMC Endocr Disord ; 21(1): 170, 2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34416890

ABSTRACT

BACKGROUND: The occurrence of hypomagnesemia in patients with primary hyperparathyroidism (PHPT) has been noted previously; however, the association of hypomagnesemia and severity of primary hyperparathyroidism remains unknown. The present study aimed to evaluate the association of hypomagnesemia with biochemical and clinical manifestations in patients with PHPT. METHODS: This was a retrospective study conducted at a tertiary hospital. We obtained data from 307 patients with PHPT from January 2010 through August 2020. Data on demographics, history, laboratory findings, bone densitometry findings, and clinical presentation and complications were collected and were compared in normal magnesium group vs hypomagnesemia group. RESULTS: Among the 307 patients with PHPT included in our study, 77 patients (33/102 [32.4%] males and 44/205 [21.5%] females) had hypomagnesemia. Mean hemoglobin levels in the hypomagnesemia group were significantly lower than those in the normal magnesium group in both males and females. In contrast, patients with hypomagnesemia had a higher mean serum calcium and parathyroid hormone than individuals with normal magnesium. The typical symptoms of PHPT, such as nephrolithiasis, bone pain/fractures, polyuria, or polydipsia, were more common in the hypomagnesemia group. In addition, patients with hypomagnesemia had a higher prevalence of osteoporosis, anemia, and hypercalcemic crisis. Even after adjusting for potential confounders, including age, sex, body mass index, estimated glomerular filtration rate, and parathyroid hormone levels, these associations remained essentially unchanged. CONCLUSION: Biochemical and clinical evidence indicates that patients with PHPT with hypomagnesemia have more severe hyperparathyroidism than those without hypomagnesemia. In addition, PHPT patients with hypomagnesemia had a higher prevalence of osteoporosis, anemia, and hypercalcemic crisis.


Subject(s)
Biomarkers/blood , Bone Density , Hypercalciuria/physiopathology , Hyperparathyroidism, Primary/pathology , Nephrocalcinosis/physiopathology , Osteoporosis/pathology , Renal Tubular Transport, Inborn Errors/physiopathology , Calcium/blood , Case-Control Studies , China/epidemiology , Female , Follow-Up Studies , Humans , Hypercalciuria/blood , Hyperparathyroidism, Primary/blood , Hyperparathyroidism, Primary/epidemiology , Male , Middle Aged , Nephrocalcinosis/blood , Osteoporosis/blood , Osteoporosis/etiology , Parathyroid Hormone/blood , Prognosis , Prospective Studies , Renal Tubular Transport, Inborn Errors/blood
3.
Int J Mol Sci ; 22(7)2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33808324

ABSTRACT

Calcium (Ca2+) is an important mediator of multicellular homeostasis and is involved in several diseases. The interplay among the kidney, bone, intestine, and parathyroid gland in Ca2+ homeostasis is strictly modulated by numerous hormones and signaling pathways. The calcium-sensing receptor (CaSR) is a G protein-coupled receptor, that is expressed in calcitropic tissues such as the parathyroid gland and the kidney, plays a pivotal role in Ca2+ regulation. CaSR is important for renal Ca2+, as a mutation in this receptor leads to hypercalciuria and calcium nephrolithiasis. In addition, CaSR is also widely expressed in the vascular system, including vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs) and participates in the process of vascular calcification. Aberrant Ca2+ sensing by the kidney and VSMCs, owing to altered CaSR expression or function, is associated with the formation of nephrolithiasis and vascular calcification. Based on emerging epidemiological evidence, patients with nephrolithiasis have a higher risk of vascular calcification, but the exact mechanism linking the two conditions is unclear. However, a dysregulation in Ca2+ homeostasis and dysfunction in CaSR might be the connection between the two. This review summarizes renal calcium handling and calcium signaling in the vascular system, with a special focus on the link between nephrolithiasis and vascular calcification.


Subject(s)
Calcium Signaling/physiology , Nephrolithiasis/metabolism , Vascular Calcification/metabolism , Animals , Calcium/metabolism , Endothelial Cells/metabolism , Humans , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Kidney/metabolism , Kidney Calculi/metabolism , Myocytes, Smooth Muscle/metabolism , Nephrolithiasis/physiopathology , Receptors, Calcium-Sensing/genetics , Vascular Calcification/genetics , Vascular Calcification/physiopathology
4.
Am J Physiol Renal Physiol ; 318(2): F363-F374, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31790303

ABSTRACT

In stone formers (SFs) with idiopathic hypercalciuria, urine pH governs the mineral phase of stones. Calcium phosphate (CaP) SFs have higher urine pH than calcium oxalate (CaOx) SFs. Normal women have higher urine pH than men on fixed diets, accompanied by greater absorption of food alkali. Female CaP and male CaOx SFs have similar urine pH as same sex normal individuals, but male CaP and female CaOx SFs may have abnormal acid-base handling. We studied 25 normal individuals (13 men and 12 women), 17 CaOx SFs (11 men and 6 women), and 15 CaP SFs (8 men and 7 women) on fixed diets. Urine and blood samples were collected under fasting and fed conditions. Female CaOx SFs had lower urine pH and lower alkali absorption, fed, compared with normal women; their urine NH4 was higher and urine citrate excretion lower than in normal women, consistent with their higher net acid excretion. Male CaOx SFs had higher urine citrate excretion and higher serum ultrafilterable citrate levels than normal men. Both male and female CaP SFs had higher urine pH fasting than same sex normal individuals, but only men were higher in the fed period, and there were no differences from normal in gut alkali absorption. CaP SFs of both sexes had higher urine NH4 and lower urine citrate than same sex normal individuals. The lower urine pH of female CaOx SFs seems related to decreased gut alkali absorption, while the higher pH of CaP SFs, accompanied by higher urine NH4 and lower urine citrate, suggests a proximal tubule disorder.


Subject(s)
Acid-Base Equilibrium , Acid-Base Imbalance/urine , Calcium Oxalate/urine , Calcium Phosphates/urine , Hypercalciuria/urine , Kidney Calculi/urine , Kidney Tubules, Proximal/metabolism , Acid-Base Imbalance/blood , Acid-Base Imbalance/diagnosis , Acid-Base Imbalance/physiopathology , Adult , Ammonium Compounds/urine , Biomarkers/blood , Biomarkers/urine , Case-Control Studies , Citric Acid/urine , Crystallization , Diet/adverse effects , Female , Gastrointestinal Absorption , Humans , Hydrogen-Ion Concentration , Hypercalciuria/blood , Hypercalciuria/diagnosis , Hypercalciuria/physiopathology , Kidney Calculi/blood , Kidney Calculi/diagnosis , Kidney Calculi/physiopathology , Kidney Tubules, Proximal/physiopathology , Male , Middle Aged , Risk Factors , Sex Factors , Young Adult
5.
BMC Nephrol ; 21(1): 171, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393202

ABSTRACT

BACKGROUND: Female Dent disease 1 patients with low-molecular-weight proteinuria (LMWP) due to CLCN5 gene mutation were rarely reported, and these cases that the people were also with Turner syndrome (TS) were even hardly documented before. CASE PRESENTATION: Here we report a 3-year and 11-month old Chinese girl with short stature who had a karyotype of 46,X,i(X)(q10) and a de novo pathogenic variant in the CLCN5 gene on the short arm of X chromosome. Laboratory examinations showed that the patient had LMWP, hypercalciuria, hypophosphatemia, delayed bone age, and genital dysplasia. CONCLUSION: The combination of i(X)(q10) and CLCN5 mutation causes the deletion of the wild-type CLCN5 allele that results in Dent-1 and TS. To the best of our knowledge, this is the first case that a female CLCN5 mutation hemizygote is diagnosed with Dent-1 and Turner syndrome due to isochromosome X. Also, our case has indicated that the prevalence of the situation may be largely underestimated because of the mild signs of females with Dent-1.


Subject(s)
Chloride Channels/genetics , Genetic Diseases, X-Linked/genetics , Nephrolithiasis/genetics , Turner Syndrome/genetics , Bone Development , Bone and Bones/diagnostic imaging , Child, Preschool , Female , Genetic Diseases, X-Linked/complications , Genetic Diseases, X-Linked/physiopathology , Hemizygote , Humans , Hypercalciuria/physiopathology , Hypophosphatemia/physiopathology , Isochromosomes , Kidney/diagnostic imaging , Mutation , Nephrolithiasis/complications , Nephrolithiasis/physiopathology , Ovary/abnormalities , Ovary/diagnostic imaging , Proteinuria/physiopathology , Turner Syndrome/complications , Turner Syndrome/physiopathology , Uterus/abnormalities , Uterus/diagnostic imaging
6.
Mediators Inflamm ; 2020: 5785378, 2020.
Article in English | MEDLINE | ID: mdl-33144848

ABSTRACT

PURPOSE: Systemic mastocytosis (SM) is characterized by a clonal proliferation of neoplastic mast cells (MCs) in one or more extracutaneous organs including the bone marrow (BM). SM is often associated with osteoporosis (OP) and fractures. Hypertryptasemia usually occurs in SM. We investigated the prevalence of hypertryptasemia in a series of severe osteoporotic patients, the performance of the tryptase test in diagnosing SM in these patients, and their bone features. METHODS: The medical records of 232 patients (168 females and 64 males) with a diagnosis of OP (50.4% with fractures) and a serum tryptase assessment were reviewed. BM assessment was performed in a subset of hypertryptasemic patients; clinical, biochemical, and radiographic data were collected. RESULTS: Hypertryptasemia was detected in 33 patients. BM assessment (n = 16) was normal in 8 hypertryptasemic patients, while BM criteria for the diagnosis of SM were met in 3 patients, MC alterations were detected in 4 patients, and one patient presented a polycythemia vera. Serum tryptase levels were higher than 11.4 ng/ml in all patients with BM alterations. The best cut-off of tryptase level related to BM alterations was 17.9 ng/ml, with a sensibility and sensitivity of 75% (AUC = 0.797 and P = 0.015 by ROC analysis). All osteoporotic patients with hypertryptasemia experienced at least one vertebral fracture associated with a severe reduction of the lumbar bone mineral density. CONCLUSIONS: The prevalence of MC-related disorders in severe OP was 3.0%, accounting for the 7.4% of the secondary causes of OP. MC-related disorders may be involved in bone fragility and assessment of serum tryptase is useful to detect MC-related disorders.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Hypercalciuria/blood , Hypercalciuria/physiopathology , Mast Cells/pathology , Mastocytosis, Systemic/pathology , Adult , Aged , Amino Acid Metabolism, Inborn Errors/blood , Bone Marrow/metabolism , Bone Marrow/pathology , Female , Humans , Male , Mastocytosis, Systemic/blood , Middle Aged , Osteoporosis/blood , Osteoporosis/physiopathology , Tryptases/metabolism
7.
Am J Physiol Renal Physiol ; 316(5): F966-F969, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30838875

ABSTRACT

The proximal tubule (PT) is responsible for the majority of calcium reabsorption by the kidney. Most PT calcium transport appears to be passive, although the molecular facilitators have not been well established. Emerging evidence supports a major role for PT calcium transport in idiopathic hypercalciuria and the development of kidney stones. This review will cover recent developments in our understanding of PT calcium transport and the role of the PT in kidney stone formation.


Subject(s)
Calcium/metabolism , Hypercalciuria/complications , Kidney Calculi/etiology , Kidney Tubules, Proximal/metabolism , Membrane Transport Proteins/metabolism , Nephrocalcinosis/etiology , Renal Reabsorption , Animals , Claudins/metabolism , Humans , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Ion Transport , Kidney Calculi/metabolism , Kidney Calculi/physiopathology , Kidney Tubules, Proximal/physiopathology , Nephrocalcinosis/metabolism , Nephrocalcinosis/physiopathology
8.
Kidney Int ; 93(3): 580-588, 2018 03.
Article in English | MEDLINE | ID: mdl-29129401

ABSTRACT

The tight junction proteins claudin-10 and -16 are crucial for the paracellular reabsorption of cations along the thick ascending limb of Henle's loop in the kidney. In patients, mutations in CLDN16 cause familial hypomagnesemia with hypercalciuria and nephrocalcinosis, while mutations in CLDN10 impair kidney function. Mice lacking claudin-16 display magnesium and calcium wasting, whereas absence of claudin-10 results in hypermagnesemia and interstitial nephrocalcinosis. In order to study the functional interdependence of claudin-10 and -16 we generated double-deficient mice. These mice had normal serum magnesium and urinary excretion of magnesium and calcium and showed polyuria and sodium retention at the expense of increased renal potassium excretion, but no nephrocalcinosis. Isolated thick ascending limb tubules of double mutants displayed a complete loss of paracellular cation selectivity and functionality. Mice lacking both claudin-10 and -16 in the thick ascending limb recruited downstream compensatory mechanisms and showed hypertrophic distal convoluted tubules with changes in gene expression and phosphorylation of ion transporters in this segment, presumably triggered by the mild decrease in serum potassium. Thus, severe individual phenotypes in claudin-10 and claudin-16 knockout mice are corrected by the additional deletion of the other claudin.


Subject(s)
Claudins/deficiency , Hypercalciuria/prevention & control , Kidney Tubules, Distal/metabolism , Loop of Henle/metabolism , Magnesium Deficiency/prevention & control , Animals , Calcium/metabolism , Claudins/genetics , Disease Models, Animal , Gene Deletion , Genetic Predisposition to Disease , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Kidney Tubules, Distal/pathology , Kidney Tubules, Distal/physiopathology , Loop of Henle/pathology , Loop of Henle/physiopathology , Magnesium/metabolism , Magnesium Deficiency/genetics , Magnesium Deficiency/metabolism , Magnesium Deficiency/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Nephrocalcinosis/genetics , Nephrocalcinosis/metabolism , Nephrocalcinosis/physiopathology , Nephrocalcinosis/prevention & control , Phenotype , Sodium/metabolism
9.
Kidney Int ; 91(4): 842-855, 2017 04.
Article in English | MEDLINE | ID: mdl-28143656

ABSTRACT

Dent disease is a rare X-linked tubulopathy caused by mutations in the endosomal chloride-proton exchanger (ClC-5) resulting in defective receptor-mediated endocytosis and severe proximal tubule dysfunction. Bone marrow transplantation has recently been shown to preserve kidney function in cystinosis, a lysosomal storage disease causing proximal tubule dysfunction. Here we test the effects of bone marrow transplantation in Clcn5Y/- mice, a faithful model for Dent disease. Transplantation of wild-type bone marrow in Clcn5Y/- mice significantly improved proximal tubule dysfunction, with decreased low-molecular-weight proteinuria, glycosuria, calciuria, and polyuria four months after transplantation, compared to Clcn5Y/- mice transplanted with ClC-5 knockout bone marrow. Bone marrow-derived cells engrafted in the interstitium, surrounding proximal tubule cells, which showed a rescue of the apical expression of ClC-5 and megalin receptors. The improvement of proximal tubule dysfunction correlated with Clcn5 gene expression in kidneys of mice transplanted with wild-type bone marrow cells. Coculture of Clcn5Y/- proximal tubule cells with bone marrow-derived cells confirmed rescue of ClC-5 and megalin, resulting in improved endocytosis. Nanotubular extensions between the engrafted bone marrow-derived cells and proximal tubule cells were observed in vivo and in vitro. No rescue was found when the formation of the tunneling nanotubes was prevented by actin depolymerization or when cells were physically separated by transwell inserts. Thus, bone marrow transplantation may rescue the epithelial phenotype due to an inherited endosomal defect. Direct contacts between bone marrow-derived cells and diseased tubular cells play a key role in the rescue mechanism.


Subject(s)
Bone Marrow Transplantation , Chloride Channels/deficiency , Dent Disease/surgery , Kidney Tubules, Proximal/physiopathology , Animals , Cell Communication , Cells, Cultured , Chloride Channels/genetics , Coculture Techniques , Dent Disease/genetics , Dent Disease/metabolism , Dent Disease/physiopathology , Disease Models, Animal , Endocytosis , Genetic Predisposition to Disease , Glycosuria/genetics , Glycosuria/metabolism , Glycosuria/physiopathology , Glycosuria/prevention & control , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Hypercalciuria/prevention & control , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Polyuria/genetics , Polyuria/metabolism , Polyuria/physiopathology , Polyuria/prevention & control , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/physiopathology , Proteinuria/prevention & control , Recovery of Function , Transplantation Chimera
10.
Physiology (Bethesda) ; 30(4): 317-26, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26136545

ABSTRACT

In addition to its prominent role in the parathyroid gland, the calcium-sensing receptor (CaSR) is expressed in various tissues, including the kidney. This article reviews current data on the calcium-sensing properties of the kidney, the localization of the CaSR protein along the nephron, and its function in calcium homeostasis and in hypercalciuria.


Subject(s)
Calcium/metabolism , Kidney Tubules/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Homeostasis , Humans , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Kidney Tubules/physiopathology , Signal Transduction
12.
Biochem Biophys Res Commun ; 456(2): 670-5, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25498502

ABSTRACT

The Na(+)/Ca(2+) exchanger (NCX) is a bidirectional transporter that is controlled by membrane potential and transmembrane gradients of Na(+) and Ca(2+). Although two isoforms of NCX1 and NCX2 are coexpressed on the basolateral membrane of the distal nephron, the functional significance of these isoforms is not entirely clear. Therefore, we used NCX1- and NCX2-heterozygote knockout mice (KO) and their double KO, as well as isoform-selective NCX inhibitors, to determine the roles of NCX isoforms in urine formation and electrolyte excretion in mice. NCX inhibitors, particularly NCX2-sensitive inhibitors, caused a dose-dependent natriuresis and in a higher dose, moreover, hypercalciuria. Consistently, NCX1-KO possessed normal renal function similar to wild-type mice (WT), whereas NCX2-KO and double KO exhibited moderate natriuresis and hypercalciuria. Notably, renal responses to YM-244769 were equivalently observed in NCX1-KO and WT, but disappeared in NCX2-KO and double KO. Thus, functional inhibition of NCX2 initially causes natriuresis, and further inhibition of NCX2 produces hypercalciuria, suggesting that the functional significance of NCX2 lies in Na(+) and Ca(2+) reabsorption of the kidney.


Subject(s)
Hypercalciuria/physiopathology , Natriuresis/physiology , Sodium-Calcium Exchanger/metabolism , Aniline Compounds/pharmacology , Animals , Gene Knockout Techniques , Hypercalciuria/genetics , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Natriuresis/drug effects , Natriuresis/genetics , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenyl Ethers/pharmacology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/genetics , Thiourea/analogs & derivatives , Thiourea/pharmacology
13.
Am J Physiol Regul Integr Comp Physiol ; 309(1): R85-92, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25947170

ABSTRACT

Idiopathic hypercalciuria (IH) is a common familial trait among patients with calcium nephrolithiasis. Previously, we have demonstrated that hypercalciuria is primarily due to reduced renal proximal and distal tubule calcium reabsorption. Here, using measurements of the clearances of sodium, calcium, and endogenous lithium taken from the General Clinical Research Center, we test the hypothesis that patterns of segmental nephron tubule calcium reabsorption differ between the sexes in IH and normal subjects. When the sexes are compared, we reconfirm the reduced proximal and distal calcium reabsorption. In IH women, distal nephron calcium reabsorption is decreased compared to normal women. In IH men, proximal tubule calcium reabsorption falls significantly, with a more modest reduction in distal calcium reabsorption compared to normal men. Additionally, we demonstrate that male IH patients have lower systolic blood pressures than normal males. We conclude that women and men differ in the way they produce the hypercalciuria of IH, with females reducing distal reabsorption and males primarily reducing proximal tubule function.


Subject(s)
Calcium/urine , Hypercalciuria/metabolism , Kidney Calculi/metabolism , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/metabolism , Renal Reabsorption , Adult , Aged , Blood Pressure , Case-Control Studies , Fasting/urine , Female , Humans , Hypercalciuria/physiopathology , Hypercalciuria/urine , Kidney Calculi/physiopathology , Kidney Calculi/urine , Kidney Tubules, Distal/physiopathology , Kidney Tubules, Proximal/physiopathology , Magnesium/urine , Male , Middle Aged , Models, Biological , Postprandial Period , Sex Factors , Sodium/urine , Time Factors , Young Adult
14.
Am J Kidney Dis ; 65(1): 122-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25446019

ABSTRACT

Loss-of-function mutations of CYP24A1, the enzyme that converts the major circulating and active forms of vitamin D to inactive metabolites, recently have been implicated in idiopathic infantile hypercalcemia. Patients with biallelic mutations in CYP24A1 present with severe hypercalcemia and nephrocalcinosis in infancy or hypercalciuria, kidney stones, and nephrocalcinosis in adulthood. We describe a cohort of 7 patients (2 adults, 5 children) presenting with severe hypercalcemia who had homozygous or compound heterozygous mutations in CYP24A1. Acute episodes of hypercalcemia in infancy were the first symptom in 6 of 7 patients; in all patients, symptoms included nephrocalcinosis, hypercalciuria, low parathyroid hormone (PTH) levels, and higher than expected 1,25-dihydroxyvitamin D levels. Longitudinal data suggested that in most patients, periods of increased sunlight exposure tended to correlate with decreases in PTH levels and increases in calcemia and calciuria. Follow-up of the 2 adult patients showed reduced glomerular filtration rate and extrarenal manifestations, including calcic corneal deposits and osteoporosis. Cases of severe PTH-independent hypercalcemia associated with hypercalciuria in infants should prompt genetic analysis of CYP24A1. These patients should be monitored carefully throughout life because they may be at increased risk for developing chronic kidney disease.


Subject(s)
Diphosphonates/pharmacology , Fluid Therapy/methods , Hypercalcemia , Nephrocalcinosis , Nephrolithiasis , Sunlight/adverse effects , Vitamin D3 24-Hydroxylase/genetics , Vitamin D/analogs & derivatives , Bone Density Conservation Agents/pharmacology , Calcium/metabolism , Child , Child, Preschool , Female , Humans , Hypercalcemia/genetics , Hypercalcemia/physiopathology , Hypercalciuria/genetics , Hypercalciuria/physiopathology , Infant , Kidney Function Tests/methods , Male , Middle Aged , Monitoring, Physiologic/methods , Mutation , Nephrocalcinosis/etiology , Nephrocalcinosis/metabolism , Nephrocalcinosis/physiopathology , Nephrolithiasis/etiology , Nephrolithiasis/metabolism , Nephrolithiasis/physiopathology , Parathyroid Hormone/blood , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/prevention & control , Seasons , Treatment Outcome , Vitamin D/metabolism , Vitamin D/pharmacology , Vitamin D3 24-Hydroxylase/metabolism
15.
Ren Fail ; 37(1): 180-3, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25366522

ABSTRACT

Familial hypomagnesemia with hypercalciuria and nephrocalcinosis is a rare autosomal recessive renal disease caused by mutations in genes for the tight junction transmembrane proteins Claudin-16 (CLDN16) and Claudin-19 (CLDN19). We present the first case report of a Mexican family with three affected sisters carrying a p.Gly20Asp mutation in CLDN19 whose heterozygous mother showed evident hypercalciuria and normal low magnesemia without any other clinical, laboratory, and radiological symptoms of renal disease making of her an unsuitable donor. The affected sisters showed variable phenotypic expression including age of first symptoms, renal urinary tract infections, nephrolithiasis, nephrocalcinosis, and eye symptoms consisting in retinochoroiditis, strabismus, macular scars, bilateral anisocoria, and severe myopia and astigmatism. End stage renal disease due to renal failure needed kidney transplantation in the three of them. Interesting findings were a heterozygous mother with asymptomatic hypercalciuria warning on the need of carefully explore clinical, laboratory, kidney ultrasonograpy, and mutation status in first degree asymptomatic relatives to avoid inappropriate kidney donors; an evident variable phenotypic expression among patients; the identification of a mutation almost confined to Spanish cases and a 3.5 Mb block of genomic homozygosis strongly suggesting a common remote parental ancestor for the gene mutation reported.


Subject(s)
Claudins/genetics , Hypercalciuria , Kidney Failure, Chronic , Nephrocalcinosis , Renal Tubular Transport, Inborn Errors , Adult , Female , Genetic Carrier Screening , Humans , Hypercalciuria/complications , Hypercalciuria/diagnosis , Hypercalciuria/ethnology , Hypercalciuria/genetics , Hypercalciuria/physiopathology , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/surgery , Kidney Transplantation , Mexico , Middle Aged , Mutation , Nephrocalcinosis/complications , Nephrocalcinosis/diagnosis , Nephrocalcinosis/ethnology , Nephrocalcinosis/genetics , Nephrocalcinosis/physiopathology , Pedigree , Renal Tubular Transport, Inborn Errors/complications , Renal Tubular Transport, Inborn Errors/diagnosis , Renal Tubular Transport, Inborn Errors/ethnology , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/physiopathology
16.
Curr Opin Nephrol Hypertens ; 23(5): 494-501, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24992569

ABSTRACT

PURPOSE OF REVIEW: The calcium-sensing receptor (CaSR) has a central role in parathyroid gland function. Genetic alterations in CaSR are well known to cause inherited forms of abnormal calcium homeostasis. This review focuses on studies investigating the role of CaSR in common disorders of abnormal calcium handling and in cardiovascular calcification. RECENT FINDINGS: Genetic population studies tested the association of common allelic CASR variants with serum and urine calcium levels, kidney stone disease, primary hyperparathyroidism and bone mineral density. The results of these association studies suggested either minor or no effects of CASR variants in these phenotypes. Decreased expression of CaSR was associated with the etiology of cardiovascular calcification in individuals with advanced chronic kidney disease. SUMMARY: Ionized calcium plays a central role in the physiology of many organ systems and disease states, but the roles of CaSR other than as illustrated by Mendelian forms of CaSR dysfunction remain unclear. The contributions of CaSR to bone mineral homeostasis, vascular calcification and other forms of cardiovascular disease need further investigation.


Subject(s)
Calcium/metabolism , Cardiovascular Diseases/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Bone Remodeling , Calcium Signaling , Cardiovascular Diseases/genetics , Cardiovascular Diseases/physiopathology , Genetic Predisposition to Disease , Homeostasis , Humans , Hypercalcemia/genetics , Hypercalcemia/metabolism , Hypercalcemia/physiopathology , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Kidney Calculi/genetics , Kidney Calculi/metabolism , Kidney Calculi/physiopathology , Mutation , Phenotype , Receptors, Calcium-Sensing/genetics , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/physiopathology
17.
Calcif Tissue Int ; 94(5): 531-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24481706

ABSTRACT

Genetic hypercalciuric stone-forming (GHS) rats, bred to maximize urine (u) calcium (Ca) excretion, demonstrate increased intestinal Ca absorption, increased bone Ca resorption, and reduced renal Ca reabsorption, all leading to elevated uCa compared to the parental Sprague-Dawley (SD) rats. GHS rats have increased numbers of vitamin D receptors (VDRs) at each site, with normal levels of 1,25(OH)2D3 (1,25D), suggesting their VDR is undersaturated with 1,25D. We have shown that 1,25D induces a greater increase in uCa in GHS than SD rats. To examine the effect of the increased VDR on the osseous response to 1,25D, we fed GHS and SD rats an ample Ca diet and injected either 1,25D [low dose (LD) 12.5 or high dose (HD) 25 ng/100 g body weight/day] or vehicle (veh) daily for 16 days. Femoral areal bone mineral density (aBMD, by DEXA) was decreased in GHS+LD and GHS+HD relative to GHS+veh, while there was no effect on SD. Vertebral aBMD was lower in GHS compared to SD and further decreased in GHS+HD. Both femoral and L6 vertebral volumetric BMD (by µCT) were lower in GHS and further reduced by HD. Histomorphometry indicated a decreased osteoclast number in GHS+HD compared to GHS+veh or SD+HD. In tibiae, GHS+HD trabecular thickness and number increased, with a 12-fold increase in osteoid volume but only a threefold increase in bone volume. Bone formation rate was decreased in GHS+HD relative to GHS+veh, confirming the mineralization defect. The loss of BMD and the mineralization defect in GHS rats contribute to increased hypercalciuria; if these effects persist, they would result in decreased bone strength, making these bones more fracture-prone. The enhanced effect of 1,25D in GHS rats indicates that the increased VDRs are biologically active.


Subject(s)
Bone Density/physiology , Calcification, Physiologic/physiology , Calcitriol/pharmacology , Hypercalciuria/physiopathology , Animals , Bone Resorption/physiopathology , Bone and Bones/drug effects , Bone and Bones/physiopathology , Calcification, Physiologic/drug effects , Calcitriol/metabolism , Disease Models, Animal , Hypercalciuria/metabolism , Male , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Receptors, Calcitriol/metabolism
18.
Ann Nutr Metab ; 64(3-4): 304-13, 2014.
Article in English | MEDLINE | ID: mdl-25300274

ABSTRACT

BACKGROUND: Idiopathic hypercalciuria (IHC), i.e. an elevated urinary calcium excretion without concomitant hypercalcemia, is a common disorder in children and can have a range of urinary clinical presentations and decreased bone mineral density (BMD). AIM: To assess the effect of IHC on bone mineral content in children without urological symptoms. METHODS: Calcium excretion, BMD (by dual-energy X-ray absorptiometry), and anthropometry were assessed in 175 seven-year-old children who were classified as IHC or controls. Calcium intake and physical activity were measured as confounding factors. RESULTS: The prevalence of IHC was 17.7%. Both groups (controls and IHC) showed similar baseline characteristics in terms of their anthropometry, gender distribution, and protein and calcium dietary intakes as well as physical activity scores. Urinary calciuria was independent of the calcium dietary intake and anthropometry. BMD correlated with anthropometry and physical activity but not with calcium dietary intake. IHC children had lower whole-body BMD z-scores compared to controls. The role of IHC in reducing the whole-body BMD z-score was still significant even when anthropometry, physical activity, and calcium intake were included as confounders in multivariate analyses. CONCLUSIONS: The prevalence of IHC in this population of 7-year-old children was about 17%. IHC diagnosis was associated with lower BMD z-scores and osteopenia in 22% of them.


Subject(s)
Bone Diseases, Developmental/etiology , Bone Diseases, Metabolic/etiology , Hypercalciuria/physiopathology , Absorptiometry, Photon , Bone Density , Calcium/urine , Calcium, Dietary/administration & dosage , Child , Child Development , Child Nutritional Physiological Phenomena , Female , Humans , Hypercalciuria/diagnostic imaging , Hypercalciuria/epidemiology , Hypercalciuria/urine , Male , Motor Activity , Osteogenesis , Prevalence , Prospective Studies , Risk , Severity of Illness Index , Spain/epidemiology , Whole Body Imaging
19.
Am J Physiol Renal Physiol ; 305(6): F853-60, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23863465

ABSTRACT

Patients with idiopathic hypercalciuria (IH) have decreased renal calcium reabsorption, most marked in the postprandial state, but the mechanisms are unknown. We compared 29 subjects with IH and 17 normal subjects (N) each fed meals providing identical amounts of calcium. Urine and blood samples were collected fasting and after meals. Levels of three candidate signalers, serum calcium (SCa), insulin (I), and plasma parathyroid hormone (PTH), did not differ between IH and N either fasting or fed, but all changed with feeding, and the change in SCa was greater in IH than in N. Regression analysis of fractional excretion of calcium (FECa) was significant for PTH and SCa in IH but not N. With the use of multivariable analysis, Sca entered the model while PTH and I did not. To avoid internal correlation we decomposed FECa into its independent terms: adjusted urine calcium (UCa) and UFCa molarity. Analyses using adjusted Uca and unadjusted Uca parallel those using FECa, showing a dominant effect of SCa with no effect of PTH or I. The effect of SCa may be mediated via vitamin D receptor-stimulated increased abundance of basolateral Ca receptor, which is supported by the fact PTH levels also seem more responsive to serum Ca in IH than in N. Although our data support an effect of SCa on FECa and UCa, which is more marked in IH than in N, it can account for only a modest fraction of the meal effect, perhaps 10-20%, suggesting additional mediators are also responsible for the exaggerated postprandial hypercalciuria seen in IH.


Subject(s)
Calcium/blood , Calcium/urine , Hypercalciuria/physiopathology , Kidney Tubules/physiology , Parathyroid Glands/physiology , Parathyroid Hormone/blood , Adult , Fasting , Female , Humans , Insulin/blood , Male , Middle Aged , Postprandial Period
20.
Am J Physiol Renal Physiol ; 304(8): F1037-42, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23364801

ABSTRACT

The study of human physiology is paramount to understanding disease and developing rational and targeted treatments. Conversely, the study of human disease can teach us a lot about physiology. Investigations into primary inherited nephrogenic diabetes insipidus (NDI) have contributed enormously to our understanding of the mechanisms of urinary concentration and identified the vasopressin receptor AVPR2, as well as the water channel aquaporin-2 (AQP2), as key players in water reabsorption in the collecting duct. Yet, there are also secondary forms of NDI, for instance as a complication of lithium treatment. The focus of this review is secondary NDI associated with inherited human diseases, such as Bartter syndrome or apparent mineralocorticoid excess. Currently, the underlying pathophysiology of this inherited secondary NDI is unclear, but there appears to be true AQP2 deficiency. To better understand the underlying mechanism(s), collaboration between clinical and experimental physiologists is essential to further investigate these observations in appropriate experimental models.


Subject(s)
Aquaporin 2/genetics , Bartter Syndrome/genetics , Bartter Syndrome/physiopathology , Diabetes Insipidus, Nephrogenic/genetics , Diabetes Insipidus, Nephrogenic/physiopathology , Receptors, Vasopressin/genetics , Aquaporin 2/deficiency , Bartter Syndrome/metabolism , Diabetes Insipidus, Nephrogenic/metabolism , Fanconi Syndrome/genetics , Fanconi Syndrome/metabolism , Fanconi Syndrome/physiopathology , Humans , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Hypokalemia/genetics , Hypokalemia/metabolism , Hypokalemia/physiopathology , Receptors, Vasopressin/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL