Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38732005

ABSTRACT

In calcium nephrolithiasis (CaNL), most calcium kidney stones are identified as calcium oxalate (CaOx) with variable amounts of calcium phosphate (CaP), where CaP is found as the core component. The nucleation of CaP could be the first step of CaP+CaOx (mixed) stone formation. High urinary supersaturation of CaP due to hypercalciuria and an elevated urine pH have been described as the two main factors in the nucleation of CaP crystals. Our previous in vivo findings (in mice) show that transient receptor potential canonical type 3 (TRPC3)-mediated Ca2+ entry triggers a transepithelial Ca2+ flux to regulate proximal tubular (PT) luminal [Ca2+], and TRPC3-knockout (KO; -/-) mice exhibited moderate hypercalciuria and microcrystal formation at the loop of Henle (LOH). Therefore, we utilized TRPC3 KO mice and exposed them to both hypercalciuric [2% calcium gluconate (CaG) treatment] and alkalineuric conditions [0.08% acetazolamide (ACZ) treatment] to generate a CaNL phenotype. Our results revealed a significant CaP and mixed crystal formation in those treated KO mice (KOT) compared to their WT counterparts (WTT). Importantly, prolonged exposure to CaG and ACZ resulted in a further increase in crystal size for both treated groups (WTT and KOT), but the KOT mice crystal sizes were markedly larger. Moreover, kidney tissue sections of the KOT mice displayed a greater CaP and mixed microcrystal formation than the kidney sections of the WTT group, specifically in the outer and inner medullary and calyceal region; thus, a higher degree of calcifications and mixed calcium lithiasis in the kidneys of the KOT group was displayed. In our effort to find the Ca2+ signaling pathophysiology of PT cells, we found that PT cells from both treated groups (WTT and KOT) elicited a larger Ca2+ entry compared to the WT counterparts because of significant inhibition by the store-operated Ca2+ entry (SOCE) inhibitor, Pyr6. In the presence of both SOCE (Pyr6) and ROCE (receptor-operated Ca2+ entry) inhibitors (Pyr10), Ca2+ entry by WTT cells was moderately inhibited, suggesting that the Ca2+ and pH levels exerted sensitivity changes in response to ROCE and SOCE. An assessment of the gene expression profiles in the PT cells of WTT and KOT mice revealed a safeguarding effect of TRPC3 against detrimental processes (calcification, fibrosis, inflammation, and apoptosis) in the presence of higher pH and hypercalciuric conditions in mice. Together, these findings show that compromise in both the ROCE and SOCE mechanisms in the absence of TRPC3 under hypercalciuric plus higher tubular pH conditions results in higher CaP and mixed crystal formation and that TRPC3 is protective against those adverse effects.


Subject(s)
Calcium Oxalate , Hypercalciuria , Kidney Calculi , Mice, Knockout , Animals , Hypercalciuria/metabolism , Hypercalciuria/genetics , Hydrogen-Ion Concentration , Mice , Calcium Oxalate/metabolism , Kidney Calculi/metabolism , Kidney Calculi/etiology , Kidney Calculi/pathology , Calcium Phosphates/metabolism , Nephrolithiasis/metabolism , Nephrolithiasis/genetics , Nephrolithiasis/pathology , Calcium/metabolism , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Disease Models, Animal , Mice, Inbred C57BL , Acetazolamide/pharmacology
2.
Curr Opin Nephrol Hypertens ; 33(4): 433-440, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38690798

ABSTRACT

PURPOSE OF REVIEW: Activation of the calcium-sensing receptor (CASR) in the parathyroid gland suppresses the release of parathyroid hormone (PTH). Furthermore, activation of the renal CASR directly increases the urinary excretion of calcium, by inhibiting transepithelial calcium transport in the nephron. Gain-of-function mutations in the CASR gene lead to autosomal dominant hypocalcemia 1 (ADH1), with inappropriately low PTH levels and hypocalcemia, indicative of excessive activation of the parathyroid CASR. However, hypercalciuria is not always observed. The reason why the manifestation of hypercalciuria is not uniform among ADH1 patients is not well understood. RECENT FINDINGS: Direct activation of the CASR in the kidney has been cumbersome to study, and an indirect measure to effectively estimate the degree of CASR activation following chronic hypercalcemia or genetic gain-of-function CASR activation has been lacking. Studies have shown that expression of the pore-blocking claudin-14 is strongly stimulated by the CASR in a dose-dependent manner. This stimulatory effect is abolished after renal Casr ablation in hypercalcemic mice, suggesting that claudin-14 abundance may gauge renal CASR activation. Using this marker has led to unexpected discoveries regarding renal CASR activation. SUMMARY: These new studies have informed on renal CASR activation thresholds and the downstream CASR-regulated calcium transport mechanisms.


Subject(s)
Kidney , Receptors, Calcium-Sensing , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/genetics , Humans , Animals , Kidney/metabolism , Hypercalciuria/metabolism , Hypercalciuria/genetics , Calcium/metabolism , Hypercalcemia/metabolism , Hypercalcemia/genetics , Claudins/metabolism , Claudins/genetics , Hypocalcemia , Hypoparathyroidism/congenital
3.
Lab Invest ; 104(5): 102047, 2024 May.
Article in English | MEDLINE | ID: mdl-38452902

ABSTRACT

Sex differences in kidney stone formation are well known. Females generally have slightly acidic blood and higher urine pH when compared with males, which makes them more vulnerable to calcium stone formation, yet the mechanism is still unclear. We aimed to examine the role of sex in stone formation during hypercalciuria and urine alkalinization through acetazolamide and calcium gluconate supplementation, respectively, for 4 weeks in wild-type (WT) and moderately hypercalciuric [TRPC3 knockout [KO](-/-)] male and female mice. Our goal was to develop calcium phosphate (CaP) and CaP+ calcium oxalate mixed stones in our animal model to understand the underlying sex-based mechanism of calcium nephrolithiasis. Our results from the analyses of mice urine, serum, and kidney tissues show that female mice (WT and KO) produce more urinary CaP crystals, higher [Ca2+], and pH in urine compared to their male counterparts. We identified a sex-based relationship of stone-forming phenotypes (types of stones) in our mice model following urine alkalization/calcium supplementation, and our findings suggest that female mice are more susceptible to CaP stones under those conditions. Calcification and fibrotic and inflammatory markers were elevated in treated female mice compared with their male counterparts, and more so in TRPC3 KO mice compared with their WT counterparts. Together these findings contribute to a mechanistic understanding of sex-influenced CaP and mixed stone formation that can be used as a basis for determining the factors in sex-related clinical studies.


Subject(s)
Hypercalciuria , Kidney Calculi , Mice, Knockout , Phenotype , Animals , Female , Male , Hypercalciuria/metabolism , Hypercalciuria/urine , Mice , Kidney Calculi/metabolism , Kidney Calculi/urine , Kidney Calculi/etiology , Calcium Phosphates/metabolism , Calcium Phosphates/urine , Hydrogen-Ion Concentration , Mice, Inbred C57BL , Disease Models, Animal , Kidney/metabolism , Sex Factors , Sex Characteristics , Calcium Oxalate/metabolism , Calcium Oxalate/urine , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics
4.
R I Med J (2013) ; 106(11): 9-13, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38015778

ABSTRACT

Idiopathic hypercalciuria is defined as excessive urine calcium excretion in the absence of an identifiable cause. It has been strongly associated with the risk of calcium kidney stone formation. Animal and human studies have suggested excessive bone mineral loss or increased gastrointestinal calcium absorption with abnormal renal calcium excretion may contribute to this process. In this article we will review the complex pathophysiology of idiopathic hypercalciuria and discuss clinical management and challenges.


Subject(s)
Hypercalciuria , Kidney Calculi , Animals , Humans , Hypercalciuria/complications , Hypercalciuria/metabolism , Calcium/metabolism , Kidney Calculi/complications , Kidney Calculi/metabolism , Kidney/metabolism
5.
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
6.
Ann N Y Acad Sci ; 1521(1): 14-31, 2023 03.
Article in English | MEDLINE | ID: mdl-36622354

ABSTRACT

Magnesium is the fourth most abundant cation in the body. It plays a critical role in many biological processes, including the process of energy release. Paracellular transport of magnesium is mandatory for magnesium homeostasis. In addition to intestinal absorption that occurs in part across the paracellular pathway, magnesium is reabsorbed by the kidney tubule. The bulk of magnesium is reabsorbed through the paracellular pathway in the proximal tubule and the thick ascending limb of the loop of Henle. The finding that rare genetic diseases due to pathogenic variants in genes encoding specific claudins (CLDNs), proteins located at the tight junction that determine the selectivity and the permeability of the paracellular pathway, led to an awareness of their importance in magnesium homeostasis. Familial hypomagnesemia with hypercalciuria and nephrocalcinosis is caused by a loss of function of CLDN16 or CLDN19. Pathogenic CLDN10 variants cause HELIX syndrome, which is associated with a severe renal loss of sodium chloride and hypermagnesemia. The present review summarizes the current knowledge of the mechanisms and factors involved in paracellular magnesium permeability. The review also highlights some of the unresolved questions that need to be addressed.


Subject(s)
Magnesium , Nephrocalcinosis , Humans , Magnesium/metabolism , Nephrocalcinosis/genetics , Nephrocalcinosis/metabolism , Hypercalciuria/genetics , Hypercalciuria/metabolism , Homeostasis , Membrane Proteins/metabolism , Claudins/genetics , Claudins/metabolism
7.
Ann Clin Lab Sci ; 52(3): 494-498, 2022 May.
Article in English | MEDLINE | ID: mdl-35777808

ABSTRACT

Autosomal dominant hypocalcemia (ADH) is characterized by hypocalcemia and inappropriately low PTH concentrations. ADH type 2 (ADH2) is caused by a heterozygous gain-of-function mutation in GNA11 that encodes the subunit of G11, the principal G protein that transduces calcium-sensing receptor signaling in the parathyroid. Clinical features related to hypocalcemia in ADH2 range from asymptomatic to tetany and seizures. We report the clinical and molecular analysis of an infant with ADH2. Exome sequencing identified a de novo heterozygous missense variant, c. G548C (p. Arg183Pro) in GNA11. This is the youngest Korean case to be diagnosed with ADH 2. In addition, we summarized the literature related to eight mutations in GNA11 from 10 families.


Subject(s)
GTP-Binding Protein alpha Subunits , Hypocalcemia , Hypoparathyroidism , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Proteins/metabolism , Humans , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypocalcemia/diagnosis , Hypocalcemia/genetics , Hypocalcemia/metabolism , Hypoparathyroidism/congenital , Hypoparathyroidism/genetics , Hypoparathyroidism/metabolism , Infant
8.
Pediatr Nephrol ; 37(4): 707-718, 2022 04.
Article in English | MEDLINE | ID: mdl-33796889

ABSTRACT

The majority of calcium filtered by the glomerulus is reabsorbed along the nephron. Most is reabsorbed from the proximal tubule (> 60%) via a paracellular pathway composed of the tight junction proteins claudins-2 and -12, a process driven by sodium and consequently water reabsorption. The thick ascending limb reabsorbs the next greatest amount of calcium (20-25%), also by a paracellular pathway composed of claudins-16 and -19. This pathway is regulated by the CaSR, whose activity increases the expression of claudin-14, a protein that blocks paracellular calcium reabsorption. The fine tuning of urinary calcium excretion occurs in the distal convoluted and connecting tubule by a transcellular pathway composed of the apical calcium channel TRPV5, the calcium shuttling protein calbindin-D28K and the basolateral proteins PMCA1b and the sodium calcium exchanger, NCX. Not surprisingly, mutations in a subset of these genes cause monogenic disorders with hypercalciuria as a part of the phenotype. More commonly, "idiopathic" hypercalciuria is encountered clinically with genetic variations in CLDN14, the CASR and TRPV5 associating with kidney stones and increased urinary calcium excretion. An understanding of the molecular pathways conferring kidney tubular calcium reabsorption is employed in this review to help explain how dietary and medical interventions for this disorder lower urinary calcium excretion.


Subject(s)
Calcium , Kidney Calculi , Calcium/metabolism , Calcium, Dietary , Claudins/genetics , Claudins/metabolism , Female , Humans , Hypercalciuria/genetics , Hypercalciuria/metabolism , Male
9.
J Am Soc Nephrol ; 32(11): 2885-2899, 2021 11.
Article in English | MEDLINE | ID: mdl-34607910

ABSTRACT

BACKGROUND: Over the last decade, advances in genetic techniques have resulted in the identification of rare hereditary disorders of renal magnesium and salt handling. Nevertheless, approximately 20% of all patients with tubulopathy lack a genetic diagnosis. METHODS: We performed whole-exome and -genome sequencing of a patient cohort with a novel, inherited, salt-losing tubulopathy; hypomagnesemia; and dilated cardiomyopathy. We also conducted subsequent in vitro functional analyses of identified variants of RRAGD, a gene that encodes a small Rag guanosine triphosphatase (GTPase). RESULTS: In eight children from unrelated families with a tubulopathy characterized by hypomagnesemia, hypokalemia, salt wasting, and nephrocalcinosis, we identified heterozygous missense variants in RRAGD that mostly occurred de novo. Six of these patients also had dilated cardiomyopathy and three underwent heart transplantation. We identified a heterozygous variant in RRAGD that segregated with the phenotype in eight members of a large family with similar kidney manifestations. The GTPase RagD, encoded by RRAGD, plays a role in mediating amino acid signaling to the mechanistic target of rapamycin complex 1 (mTORC1). RagD expression along the mammalian nephron included the thick ascending limb and the distal convoluted tubule. The identified RRAGD variants were shown to induce a constitutive activation of mTOR signaling in vitro. CONCLUSIONS: Our findings establish a novel disease, which we call autosomal dominant kidney hypomagnesemia (ADKH-RRAGD), that combines an electrolyte-losing tubulopathy and dilated cardiomyopathy. The condition is caused by variants in the RRAGD gene, which encodes Rag GTPase D; these variants lead to an activation of mTOR signaling, suggesting a critical role of Rag GTPase D for renal electrolyte handling and cardiac function.


Subject(s)
Cardiomyopathy, Dilated/genetics , Hypercalciuria/genetics , Kidney Diseases/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation, Missense , Nephrocalcinosis/genetics , Renal Tubular Transport, Inborn Errors/genetics , TOR Serine-Threonine Kinases/metabolism , Cardiomyopathy, Dilated/metabolism , Female , HEK293 Cells , Humans , Hypercalciuria/metabolism , Kidney Diseases/metabolism , Kidney Tubules, Distal/metabolism , Male , Models, Molecular , Natriuresis/genetics , Nephrocalcinosis/metabolism , Pedigree , Protein Conformation , Renal Tubular Transport, Inborn Errors/metabolism , Seizures/genetics , Seizures/metabolism , Signal Transduction , Exome Sequencing , Whole Genome Sequencing
10.
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
11.
Am J Physiol Renal Physiol ; 320(3): F418-F428, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33522409

ABSTRACT

Urinary calcium and magnesium wasting is a characteristic feature of metabolic acidosis, and this study focused on the role of the thick ascending limb of Henle's loop in metabolic acidosis-induced hypercalciuria and hypermagnesiuria because thick ascending limb is an important site of paracellular calcium and magnesium reabsorption. Male Sprague-Dawley rats were used to determine the effects of acid loading (by adding NH4Cl, 7.2 mmol/220 g body wt/day to food slurry for 7 days) on renal expression of claudins and then to evaluate whether the results were reversed by antagonizing calcium-sensing receptor (using NPS-2143). At the end of each animal experiment, the kidneys were harvested for immunoblotting, immunofluorescence microscopy, and quantitative PCR (qPCR) analysis of claudins and the calcium-sensing receptor. As expected, NH4Cl loading lowered urinary pH and increased excretion of urinary calcium and magnesium. In NH4Cl-loaded rats, renal protein and mRNA expression of claudin-16, and claudin-19, were decreased compared with controls. However, claudin-14 protein and mRNA increased in NH4Cl-loaded rats. Consistently, the calcium-sensing receptor protein and mRNA were up-regulated in NH4Cl-loaded rats. All these changes were reversed by NPS-2143 coadministration and were confirmed using immunofluorescence microscopy. Hypercalciuria and hypermagnesiuria in NH4Cl-loaded rats were significantly ameliorated by NPS-2143 coadministration as well. We conclude that in metabolic acidosis, claudin-16 and claudin-19 in the thick ascending limb are down-regulated to produce hypercalciuria and hypermagnesiuria via the calcium-sensing receptor.NEW & NOTEWORTHY This study found that the thick ascending limb of Henle's loop is involved in the mechanisms of hypercalciuria and hypermagnesiuria in metabolic acidosis. Specifically, expression of claudin-16/19 and claudin-14 was altered via up-regulation of calcium-sensing receptor in NH4Cl-induced metabolic acidosis. Our novel findings contribute to understanding the regulatory role of paracellular tight junction proteins in the thick ascending limb.


Subject(s)
Calcium/metabolism , Claudins/metabolism , Hypercalciuria/metabolism , Loop of Henle/metabolism , Magnesium/metabolism , Acidosis/metabolism , Animals , Calcium, Dietary/metabolism , Hypercalciuria/pathology , Loop of Henle/pathology , Male , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/metabolism
12.
J Clin Endocrinol Metab ; 106(4): e1775-e1792, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33340048

ABSTRACT

CONTEXT: The calcium-sensing receptor (CaSR) is essential to maintain a stable calcium concentration in serum. Spermatozoa are exposed to immense changes in concentrations of CaSR ligands such as calcium, magnesium, and spermine during epididymal maturation, in the ejaculate, and in the female reproductive environment. However, the role of CaSR in human spermatozoa is unknown. OBJECTIVE: This work aimed to investigate the role of CaSR in human spermatozoa. METHODS: We identified CaSR in human spermatozoa and characterized the response to CaSR agonists on intracellular calcium, acrosome reaction, and 3',5'-cyclic adenosine 5'-monophosphate (cAMP) in spermatozoa from men with either loss-of-function or gain-of-function mutations in CASR and healthy donors. RESULTS: CaSR is expressed in human spermatozoa and is essential for sensing extracellular free ionized calcium (Ca2+) and Mg2+. Activators of CaSR augmented the effect of sperm-activating signals such as the response to HCO3- and the acrosome reaction, whereas spermatozoa from men with a loss-of-function mutation in CASR had a diminished response to HCO3-, lower progesterone-mediated calcium influx, and were less likely to undergo the acrosome reaction in response to progesterone or Ca2+. CaSR activation increased cAMP through soluble adenylyl cyclase (sAC) activity and increased calcium influx through CatSper. Moreover, external Ca2+ or Mg2+ was indispensable for HCO3- activation of sAC. Two male patients with a CASR loss-of-function mutation in exon 3 presented with normal sperm counts and motility, whereas a patient with a loss-of-function mutation in exon 7 had low sperm count, motility, and morphology. CONCLUSION: CaSR is important for the sensing of Ca2+, Mg2+, and HCO3- in spermatozoa, and loss-of-function may impair male sperm function.


Subject(s)
Bicarbonates/metabolism , Calcium/metabolism , Receptors, Calcium-Sensing/physiology , Spermatozoa/metabolism , Acrosome Reaction/drug effects , Acrosome Reaction/genetics , Adult , Bicarbonates/pharmacology , Calcium/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/genetics , Case-Control Studies , Female , Humans , Hypercalcemia/congenital , Hypercalcemia/genetics , Hypercalcemia/metabolism , Hypercalcemia/pathology , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/pathology , Hypocalcemia/genetics , Hypocalcemia/metabolism , Hypocalcemia/pathology , Hypoparathyroidism/congenital , Hypoparathyroidism/genetics , Hypoparathyroidism/metabolism , Hypoparathyroidism/pathology , Kidney/metabolism , Kidney/pathology , Magnesium/metabolism , Magnesium/pharmacology , Male , Mutation , Receptors, Calcium-Sensing/genetics , Sperm Motility/drug effects , Sperm Motility/genetics , Spermatozoa/drug effects , Spermatozoa/physiology , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology
13.
BMC Nephrol ; 21(1): 328, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32758178

ABSTRACT

BACKGROUND: Gitelman syndrome is a rare salt-losing renal tubular disorder associated with mutation of SLC12A3 gene, which encodes the Na-Cl co-transporter (NCCT). Gitelman syndrome is characterized by hypokalemia, metabolic alkalosis, hypomagnesemia, hypocalciuria, and renin-angiotensin-aldosterone system (RAAS) activation. Different SLC12A3 variants may lead to phenotypic variability and severity. METHODS: In this study, we reported the clinical features and genetic analysis of a Chinese pedigree diagnosed with Gitelman syndrome. RESULTS: The proband exhibited hypokalaemia, hypomagnesemia, metabolic alkalosis, but hypercalciuria and kidney stone formation. The increased urinary calcium excretion made it confused to Bartter syndrome. The persistent renal potassium wasting resulted in renal tubular lesions, and might affect urinary calcium reabsorption and excretion. Genetic analysis revealed mutations of SLC12A3 gene with c.433C > T (p.Arg145Cys), c.1077C > G (p.Asn359Lys), and c.1666C > T (p.Pro556Ser). Potential alterations of structure and function of NCCT protein due to those genetic variations of SLC12A3 are predicted. Interestingly, one sibling of the proband carried the same mutant sites and exhibited similar clinical features with milder phenotypes of hypokalemia and hypomagnesemia, but hypocalciuria rather than hypercalciuria. Family members with at least one wild type copy of SLC12A3 had normal biochemistry. With administration of spironolactone, potassium chloride and magnesium supplement, the serum potassium and magnesium were maintained within normal ranges. CONCLUSIONS: In this study, we identified compound mutations of SLC12A3 associated with varieties of clinical features. Further efforts are needed to investigate the diversity in clinical manifestations of Gitelman syndrome and its correlation with specific SLC12A3 mutations.


Subject(s)
Gitelman Syndrome/genetics , Adult , Aged , Alkalosis/genetics , Alkalosis/metabolism , Bartter Syndrome/metabolism , China , Female , Genotype , Gitelman Syndrome/metabolism , Humans , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypokalemia/genetics , Hypokalemia/metabolism , Magnesium/blood , Male , Middle Aged , Mutation , Pedigree , Phenotype , Renal Elimination , Solute Carrier Family 12, Member 3/genetics , Water-Electrolyte Imbalance/genetics , Water-Electrolyte Imbalance/metabolism
14.
Nutrients ; 12(9)2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32825353

ABSTRACT

Vitamin D supplementation in patients with urolithiasis and hypercalciuria is considered to be unsafe. We analyzed the impact of vitamin D supplementation on selected health status parameters in children with idiopathic hypercalciuria. The study included 36 children with urolithiasis resulting from excessive calcium excretion. The level of calcium and 25(OH)D (hydroxylated vitamin D - calcidiol) in serum, urinary calcium excretion and the presence of stones in urinary tract were assessed prospectively. Blood and urine samples were collected at the time when the patient was qualified for the study and every three months up to 24 month of vitamin D intake at a dose of 400 or 800 IU/day. At time zero and at 12, and 24 months of vitamin D supplementation, densitometry was performed. Supplementation with vitamin D caused a statistically significant increase in the concentration of 25(OH)D in serum. There were no significant changes in calcium concentration in serum, excretion of calcium in urine but also in bone density. There was no significant increase in the risk of formation or development of stones in the urinary tract. Supplementation with vitamin D (400-800 IU/day) in children with idiopathic hypercalciuria significantly increases 25(OH)D concentration, does not affect calciuria, but also does not improve bone density.


Subject(s)
Bone Density/drug effects , Child Nutritional Physiological Phenomena/physiology , Dietary Supplements , Hypercalciuria/metabolism , Negative Results , Urinary Tract/metabolism , Urolithiasis/etiology , Vitamin D/adverse effects , Vitamin D/pharmacology , Adolescent , Child , Child, Preschool , Female , Humans , Hypercalciuria/complications , Male , Vitamin D/administration & dosage , Vitamin D/analogs & derivatives , Vitamin D/blood
15.
Nefrología (Madrid) ; 40(2): 190-196, mar.-abr. 2020. tab
Article in Spanish | IBECS | ID: ibc-199107

ABSTRACT

Existe controversia si la hipercalciuria idiopática (HI) produce alteraciones en el manejo renal del agua. Por primera vez en la literatura, llevamos a cabo un estudio longitudinal del manejo renal del agua (MRA) en pacientes diagnosticados de HI en edad pediátrica y con seguimiento hasta la edad adulta (media de seguimiento de 17,7 ± 1,4 años). MÉTODOS: Veintinueve pacientes (7 M, 22 F) mayores de 24 años (media 28,2 ± 2,9 años, rango: 24,1-35,9) que fueron diagnosticados de HI en la edad pediátrica (media 7,6 ± 3,2 años, rango: 1-14) fueron incluidos. Se determinaron la osmolaridad urinaria máxima (OsU) y/o el volumen urinario ajustado para 100 ml de tasa de filtrado glomerular (V/TFG) en ambos tiempos (pediátrico y adulto). Además, siempre que fue posible, en ambas edades se recogieron los niveles plasmáticos de creatinina, sodio plasmático, ácido úrico, cociente citrato/creatinina y calcio/citrato y, además, se realizó una ecografía renovesical. RESULTADOS: El MRA estuvo alterado en edad pediátrica en 9/29 casos (31%) (4 con OsU máxima reducida y 5 con V/TFG elevado). En la edad adulta, 7/29 (24,1%) presentaron alteración del MRA (6 OsU reducidos y uno con V/TFG elevado). En comparación con el grupo de edad pediátrica, los pacientes adultos mostraron valores reducidos de V/TFG, cociente calcio/creatinina y citrato/creatinina, así como aumento de creatinina plasmática, ácido úrico y del cociente calcio/citrato. No hubo diferencias en la OsU máxima en ambos tiempos. Sin embargo, la OsU en la edad adulta fue significativamente menor en aquellos que tenían cólicos renales comparado con aquellos que no los tuvieron (p = 0,04). CONCLUSIONES: La alteración del MRA ocurrió en aproximadamente un tercio de los pacientes con HI, y no se alteró tras 20 años después de su diagnóstico. Nosotros pensamos que estos resultados pueden ser debido a un cierto cumplimiento de la dieta protectora recomendada y al tratamiento farmacológico administrado en el diagnóstico de HI en la edad pediátrica


INTRODUCTION: There is much debate about whether idiopathic hypercalciuria (IH) affects kidney water management. For the first time in the literature, we carried out a longitudinal study of kidney water management (KWM) in patients diagnosed with IH in childhood and followed-up until adulthood (mean follow-up 17.7 ± 1.4 years). Methods; Twenty-nine patients (7 M, 22 F) over the age of 24 years (mean 28.2 ± 2.9 years, range: 24.1-35.9) who were diagnosed with IH in childhood (mean 7.6 ± 3.2 years, range: 1-14) were included. Maximum urine osmolality (UO) and/or urine volume adjusted for 100ml of glomerular filtration rate (V/GFR) in both age groups (paediatric and adult) were determined. Moreover, whenever possible, in both age groups plasma creatinine levels, plasma sodium levels, uric acid levels, the citrate/creatinine ratio and the calcium/citrate ratio were recorded and a renal and bladder ultrasound was performed. RESULTS: In the paediatric age group, KWM was altered in 9/29 cases (31%) (4 with reduced maximum UO and 5 with elevated V/GFR). In adulthood, KWM was found to be affected in 7/29 cases (24.1%) (6 with reduced UO and one with elevated V/GFR). Compared to the paediatric age group, adult patients had lower V/GFR, calcium/creatinine and citrate/creatinine values, as well as higher plasma creatinine, uric acid and calcium/citrate. There were no differences in the maximum UO in both age groups. However, UO in adulthood was significantly lower in subjects who had renal colic compared to those who did not (P = .04). CONCLUSIONS: KWM was affected in approximately one third of patients with IH, which persisted 20 years after diagnosis. We think that these results may be due to adherence to the recommended protective diet and to the pharmacological treatment administered at the diagnosis of IH during childhood


Subject(s)
Humans , Male , Female , Infant , Child, Preschool , Child , Adolescent , Adult , Hypercalciuria/metabolism , Kidney/metabolism , Water/metabolism , Antidiuretic Agents/administration & dosage , Antidiuretic Agents/urine , Citric Acid/blood , Creatinine/blood , Deamino Arginine Vasopressin/administration & dosage , Deamino Arginine Vasopressin/urine , Glomerular Filtration Rate , Hypercalciuria/blood , Longitudinal Studies , Osmolar Concentration , Sodium/blood , Uric Acid/blood , Urine/chemistry
16.
Respiration ; 99(3): 207-212, 2020.
Article in English | MEDLINE | ID: mdl-32114588

ABSTRACT

BACKGROUND: Calcium metabolism alterations are quite common in sarcoidosis and have been correlated with disease activity. OBJECTIVES: The aim of the study was to investigate the clinical significance of calcium metabolism alterations in patients with chronic sarcoidosis. We paid particular attention to associations with specific disease phenotypes and chitotriosidase (CTO) expression. METHODS: 212 chronic sarcoidosis patients (mean age 56.07 ± 12 years; 97 males) were retrospectively recruited. Demographic, clinical, functional, and radiological data, and serum-urinary calcium metabolism were entered into an electronical database for analysis. Levels of CTO and angiotensin-converting enzyme (ACE) were measured and bone mineral density and lung function tests were conducted. RESULTS: Hypercalciuria and hypercalcemia were observed in 18.8 and 1.8% of patients, respectively. Urinary calcium levels correlated with CTO activity (r = 0.33, p = 0.0042). Patients with worsening persistent disease showed the highest levels of urinary calcium. Diffusing capacity of the lung for carbon monoxide (DLCO) percentage correlated inversely with urinary calcium (r = 0.1482; p = 0.0397). CONCLUSIONS: Calcium metabolism alteration, particularly hypercalciuria, was observed in a significant percentage of patients of sarcoidosis. Urinary calcium was correlated with clinical status, DLCO, and serum CTO activity, suggesting its potential role as a biomarker of the activity and severity of sarcoidosis.


Subject(s)
Calcium/metabolism , Hexosaminidases/blood , Hypercalcemia/metabolism , Hypercalciuria/metabolism , Peptidyl-Dipeptidase A/blood , Sarcoidosis, Pulmonary/metabolism , Absorptiometry, Photon , Adult , Aged , Bone Density , Creatinine/metabolism , Female , Forced Expiratory Volume , Humans , Male , Middle Aged , Phosphates/metabolism , Pulmonary Diffusing Capacity , Radiography, Thoracic , Respiratory Function Tests , Retrospective Studies , Sarcoidosis/metabolism , Sarcoidosis/physiopathology , Sarcoidosis, Pulmonary/diagnostic imaging , Sarcoidosis, Pulmonary/physiopathology , Vital Capacity
17.
Physiol Rep ; 8(1): e14316, 2020 01.
Article in English | MEDLINE | ID: mdl-31908154

ABSTRACT

Calcineurin inhibitors (CNIs) are immunosuppressive drugs used to prevent graft rejection after organ transplant. Common side effects include renal magnesium wasting and hypomagnesemia, which may contribute to new-onset diabetes mellitus, and hypercalciuria, which may contribute to post-transplant osteoporosis. Previous work suggested that CNIs reduce the abundance of key divalent cation transport proteins, expressed along the distal convoluted tubule, causing renal magnesium and calcium wasting. It has not been clear, however, whether these effects are specific for the distal convoluted tubule, and whether these represent off-target toxic drug effects, or result from inhibition of calcineurin. The CNI tacrolimus can inhibit calcineurin only when it binds with the immunophilin, FKBP12; we previously generated mice in which FKBP12 could be deleted along the nephron, to test whether calcineurin inhibition is involved, these mice are normal at baseline. Here, we confirmed that tacrolimus-treated control mice developed hypomagnesemia and urinary calcium wasting, with decreased protein and mRNA abundance of key magnesium and calcium transport proteins (NCX-1 and Calbindin-D28k ). However, qPCR also showed decreased mRNA expression of NCX-1 and Calbindin-D28k , and TRPM6. In contrast, KS-FKBP12-/- mice treated with tacrolimus were completely protected from these effects. These results indicate that tacrolimus affects calcium and magnesium transport along the distal convoluted tubule and strongly suggests that inhibition of the phosphatase, calcineurin, is directly involved.


Subject(s)
Calcineurin Inhibitors/pharmacology , Calcium/metabolism , Hypercalciuria/chemically induced , Kidney Tubules, Distal/drug effects , Magnesium/metabolism , Tacrolimus Binding Protein 1A/genetics , Tacrolimus/pharmacology , Water-Electrolyte Imbalance/chemically induced , Animals , Calbindin 1/drug effects , Calbindin 1/genetics , Calbindin 1/metabolism , Calcineurin Inhibitors/adverse effects , Calcium/urine , Gene Expression , Hypercalciuria/metabolism , Hypercalciuria/urine , Kidney Tubules, Distal/metabolism , Magnesium/urine , Mice , Mice, Knockout , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Sodium-Calcium Exchanger/drug effects , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism , TRPM Cation Channels/drug effects , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Tacrolimus/adverse effects , Tacrolimus Binding Protein 1A/metabolism , Water-Electrolyte Imbalance/metabolism , Water-Electrolyte Imbalance/urine
18.
Pediatr Nephrol ; 35(3): 383-397, 2020 03.
Article in English | MEDLINE | ID: mdl-30607567

ABSTRACT

BACKGROUND: The incidence of nephrolithiasis in children and adolescents is increasing and appears to double every 10 years. The most important role of the pediatric nephrologist is to diagnose and modify various metabolic and non-metabolic risk factors, as well as prevent long-term complications especially in the case of recurrent nephrolithiasis. OBJECTIVE: The purpose of this review is to summarize the existing literature on the etiology and management of pediatric nephrolithiasis. RESULTS: The incidence of kidney stones is increasing; dietary and environmental factors are probably the main causes for this increased incidence. In most pediatric patients, the etiology for the kidney stones can be identified. Metabolic factors, such as hypercalciuria and hypocitraturia, urinary tract infection, and urinary stasis, constitute leading causes. Herein, we review the etiologies, diagnostic work-up, and treatment options for the most prevalent causes of kidney stones. The detrimental effects of excessive dietary sodium, reduced fluid intake, and the benefits of plant-based over animal-based protein consumption on urinary crystal formation are discussed. We also review the long-term complications. CONCLUSIONS: Pediatric nephrologists have an important role in the diagnostic work-up and prevention of recurring nephrolithiasis.


Subject(s)
Hypercalciuria/diagnosis , Hyperoxaluria/diagnosis , Kidney Calculi/diagnosis , Nephrologists/organization & administration , Professional Role , Adolescent , Child , Humans , Hypercalciuria/metabolism , Hypercalciuria/therapy , Hypercalciuria/urine , Hyperoxaluria/metabolism , Hyperoxaluria/therapy , Hyperoxaluria/urine , Incidence , Kidney Calculi/epidemiology , Kidney Calculi/metabolism , Kidney Calculi/therapy , Recurrence , Risk Factors , Secondary Prevention/organization & administration
19.
Nefrologia (Engl Ed) ; 40(2): 190-196, 2020.
Article in English, Spanish | MEDLINE | ID: mdl-31806292

ABSTRACT

INTRODUCTION: There is much debate about whether idiopathic hypercalciuria (IH) affects kidney water management. For the first time in the literature, we carried out a longitudinal study of kidney water management (KWM) in patients diagnosed with IH in childhood and followed-up until adulthood (mean follow-up 17.7±1.4 years). METHODS: Twenty-nine patients (7 M, 22 F) over the age of 24 years (mean 28.2±2.9 years, range: 24.1-35.9) who were diagnosed with IH in childhood (mean 7.6±3.2 years, range: 1-14) were included. Maximum urine osmolality (UO) and/or urine volume adjusted for 100ml of glomerular filtration rate (V/GFR) in both age groups (paediatric and adult) were determined. Moreover, whenever possible, in both age groups plasma creatinine levels, plasma sodium levels, uric acid levels, the citrate/creatinine ratio and the calcium/citrate ratio were recorded and a renal and bladder ultrasound was performed. RESULTS: In the paediatric age group, KWM was altered in 9/29 cases (31%) (4 with reduced maximum UO and 5 with elevated V/GFR). In adulthood, KWM was found to be affected in 7/29 cases (24.1%) (6 with reduced UO and one with elevated V/GFR). Compared to the paediatric age group, adult patients had lower V/GFR, calcium/creatinine and citrate/creatinine values, as well as higher plasma creatinine, uric acid and calcium/citrate. There were no differences in the maximum UO in both age groups. However, UO in adulthood was significantly lower in subjects who had renal colic compared to those who did not (P=.04). CONCLUSIONS: KWM was affected in approximately one third of patients with IH, which persisted 20 years after diagnosis. We think that these results may be due to adherence to the recommended protective diet and to the pharmacological treatment administered at the diagnosis of IH during childhood.


Subject(s)
Hypercalciuria/metabolism , Kidney/metabolism , Water/metabolism , Adolescent , Adult , Age Factors , Antidiuretic Agents/administration & dosage , Antidiuretic Agents/urine , Child , Child, Preschool , Citric Acid/blood , Creatinine/blood , Deamino Arginine Vasopressin/administration & dosage , Deamino Arginine Vasopressin/urine , Female , Glomerular Filtration Rate , Humans , Hypercalciuria/blood , Infant , Longitudinal Studies , Male , Osmolar Concentration , Sodium/blood , Uric Acid/blood , Urine/chemistry
20.
Cell Physiol Biochem ; 52(5): 1166-1177, 2019.
Article in English | MEDLINE | ID: mdl-30990586

ABSTRACT

BACKGROUND/AIMS: Tributyltin (TBT) is an organotin (OTs) and biohazard organometallic pollutant. Recently our group has shown that TBT, even in very low doses, has deleterious effects on several tissues most likely due to its role as an endocrine-disrupting molecule. Other studies have confirmed that OT exposure could be responsible for neural, endocrine, and reproductive dysfunctions via in vitro and in vivo models. However, TBT effects on bone lack concise data despite the fact that bone turnover is regulated by endocrine molecules, such as parathormone (PTH), estrogen (E2), etc. Our group has already shown that TBT disrupts adrenal and female gonadal functions. METHODS: We studied the effects of TBT on bone metabolism and structure using DXA, microCT scan, and SEM. We also determined the calcium (Ca²âº) and phosphate (Pi) metabolism in TBT-treated rats as well as some biomarkers for bone formation and resorption. RESULTS: Surprisingly, we found that TBT leads to higher bone mineral density (BMD) although lesions in spinal bone were observed by either microCT scan or SEM. Biomarkers for bone resorption, such as the urinary deoxipyridinolines (DPD) excretion ratio was increased in TBT-treated animals versus mock-treated controls. Osteocalcin (OC) and alkaline phosphatase (AP) are markers of bone formation and are also elevated suggesting that the bone matrix suffers from a higher turnover. Serum Ca²âº (total and ionized) do not changed by TBT treatment although hypercalciuria is observed. CONCLUSION: It is known that Sn atoms have three valence states (Sn²âº, Sn³âº, and Sn4⁺); hence, we hypothesized that Sn (more likely Sn²âº) could be competing with Ca²âº and/or Mg²âº in hydroxyapatite mineral matrix to disturb bone turnover. Further work is needed to confirm this hypothesis.


Subject(s)
Bone Density/drug effects , Bone Resorption , Endocrine Disruptors/toxicity , Hypercalciuria , Osteogenesis/drug effects , Trialkyltin Compounds/toxicity , Animals , Bone Resorption/chemically induced , Bone Resorption/diagnostic imaging , Bone Resorption/metabolism , Female , Hypercalciuria/chemically induced , Hypercalciuria/diagnostic imaging , Hypercalciuria/metabolism , Rats , Rats, Wistar , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL
...