Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 77
1.
Curr Opin Nephrol Hypertens ; 32(4): 359-365, 2023 07 01.
Article En | MEDLINE | ID: mdl-37074688

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.


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 En | MEDLINE | ID: mdl-34416890

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.


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 En | MEDLINE | ID: mdl-33808324

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.


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
5.
Mediators Inflamm ; 2020: 5785378, 2020.
Article En | MEDLINE | ID: mdl-33144848

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.


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
6.
BMC Nephrol ; 21(1): 171, 2020 05 11.
Article En | MEDLINE | ID: mdl-32393202

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.


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
7.
Am J Physiol Renal Physiol ; 318(2): F363-F374, 2020 02 01.
Article En | MEDLINE | ID: mdl-31790303

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.


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
8.
Am J Physiol Renal Physiol ; 316(5): F966-F969, 2019 05 01.
Article En | MEDLINE | ID: mdl-30838875

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.


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
9.
Nat Rev Endocrinol ; 15(1): 33-51, 2018 12.
Article En | MEDLINE | ID: mdl-30443043

The Ca2+-sensing receptor (CaSR) is a dimeric family C G protein-coupled receptor that is expressed in calcitropic tissues such as the parathyroid glands and the kidneys and signals via G proteins and ß-arrestin. The CaSR has a pivotal role in bone and mineral metabolism, as it regulates parathyroid hormone secretion, urinary Ca2+ excretion, skeletal development and lactation. The importance of the CaSR for these calcitropic processes is highlighted by loss-of-function and gain-of-function CaSR mutations that cause familial hypocalciuric hypercalcaemia and autosomal dominant hypocalcaemia, respectively, and also by the fact that alterations in parathyroid CaSR expression contribute to the pathogenesis of primary and secondary hyperparathyroidism. Moreover, the CaSR is an established therapeutic target for hyperparathyroid disorders. The CaSR is also expressed in organs not involved in Ca2+ homeostasis: it has noncalcitropic roles in lung and neuronal development, vascular tone, gastrointestinal nutrient sensing, wound healing and secretion of insulin and enteroendocrine hormones. Furthermore, the abnormal expression or function of the CaSR is implicated in cardiovascular and neurological diseases, as well as in asthma, and the CaSR is reported to protect against colorectal cancer and neuroblastoma but increase the malignant potential of prostate and breast cancers.


Calcimimetic Agents/therapeutic use , Hypercalcemia/congenital , Hypercalciuria/genetics , Hypocalcemia/genetics , Hypoparathyroidism/congenital , Nephrolithiasis/genetics , Receptors, Calcium-Sensing/genetics , Female , Gene Expression Regulation , Genetic Predisposition to Disease/epidemiology , Humans , Hypercalcemia/drug therapy , Hypercalcemia/genetics , Hypercalcemia/physiopathology , Hypercalciuria/drug therapy , Hypercalciuria/physiopathology , Hypocalcemia/drug therapy , Hypocalcemia/physiopathology , Hypoparathyroidism/drug therapy , Hypoparathyroidism/genetics , Hypoparathyroidism/physiopathology , Incidence , Male , Mutation/genetics , Nephrolithiasis/drug therapy , Nephrolithiasis/physiopathology , Prognosis , Receptors, Calcium-Sensing/drug effects , Risk Assessment , Treatment Outcome
10.
Sci Signal ; 11(518)2018 02 20.
Article En | MEDLINE | ID: mdl-29463778

The calcium-sensing receptor (CaSR) is a G protein-coupled receptor (GPCR) that signals through Gq/11 and Gi/o to stimulate cytosolic calcium (Ca2+i) and mitogen-activated protein kinase (MAPK) signaling to control extracellular calcium homeostasis. Studies of loss- and gain-of-function CASR mutations, which cause familial hypocalciuric hypercalcemia type 1 (FHH1) and autosomal dominant hypocalcemia type 1 (ADH1), respectively, have revealed that the CaSR signals in a biased manner. Thus, some mutations associated with FHH1 lead to signaling predominantly through the MAPK pathway, whereas mutations associated with ADH1 preferentially enhance Ca2+i responses. We report a previously unidentified ADH1-associated R680G CaSR mutation, which led to the identification of a CaSR structural motif that mediates biased signaling. Expressing CaSRR680G in HEK 293 cells showed that this mutation increased MAPK signaling without altering Ca2+i responses. Moreover, this gain of function in MAPK activity occurred independently of Gq/11 and Gi/o and was mediated instead by a noncanonical pathway involving ß-arrestin proteins. Homology modeling and mutagenesis studies showed that the R680G CaSR mutation selectively enhanced ß-arrestin signaling by disrupting a salt bridge formed between Arg680 and Glu767, which are located in CaSR transmembrane domain 3 and extracellular loop 2, respectively. Thus, our results demonstrate CaSR signaling through ß-arrestin and the importance of the Arg680-Glu767 salt bridge in mediating signaling bias.


Cell Membrane/metabolism , Hypercalciuria/physiopathology , Hypocalcemia/physiopathology , Hypoparathyroidism/congenital , MAP Kinase Signaling System , Mutation , Receptors, Calcium-Sensing/metabolism , Salts/metabolism , beta-Arrestins/metabolism , Amino Acid Sequence , Base Sequence , Calcium/metabolism , Cell Membrane/chemistry , Family Health , Female , Humans , Hypercalciuria/genetics , Hypocalcemia/genetics , Hypoparathyroidism/genetics , Hypoparathyroidism/physiopathology , Male , Models, Molecular , Pedigree , Protein Conformation , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/genetics , Salts/chemistry , Sequence Homology, Amino Acid
11.
Kidney Int ; 93(3): 580-588, 2018 03.
Article En | MEDLINE | ID: mdl-29129401

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.


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
13.
Iran J Kidney Dis ; 11(3): 180-191, 2017 May.
Article En | MEDLINE | ID: mdl-28575878

The pathophysiology of urinary stone formation is complex, involving a combination of metabolic, genetic, and environmental factors. Over the past decades, remarkable advances have been emerged in the understanding of the pathogenesis, diagnosis, and treatment of calcium kidney calculi. For this review, both original and review articles were found via PubMed search on pathophysiology, diagnosis, and management of urinary calculi. These resources were integrated with the authors' knowledge of the field. Nephrolithiasis is suggested to be associated with systemic disorders, including chronic kidney insufficiency, hematologic malignancies, endocrine disorders, autoimmune diseases, inflammatory bowel diseases, bone loss and fractures, hypertension, type 2 diabetes mellitus, metabolic syndrome, and vascular diseases like coronary heart diseases and most recently ischemic strokes. This is changing the perspective of nephrolithiasis from an isolated disorder to a systemic disease that justifies further research in understanding the underlying mechanisms and elaborating diagnostic-therapeutic options.


Hypercalciuria , Kidney Calculi , Kidney , Animals , Humans , Hypercalciuria/diagnosis , Hypercalciuria/epidemiology , Hypercalciuria/physiopathology , Hypercalciuria/therapy , Kidney/physiopathology , Kidney Calculi/diagnosis , Kidney Calculi/epidemiology , Kidney Calculi/physiopathology , Kidney Calculi/therapy , Prognosis , Risk Factors
14.
Kidney Int ; 91(4): 842-855, 2017 04.
Article En | MEDLINE | ID: mdl-28143656

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.


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
15.
Nefrología (Madr.) ; 37(1): 5-8, ene.-feb. 2017. ilus
Article Es | IBECS | ID: ibc-160592

La reabsorción tubular de calcio es uno de los principales factores que determinan la concentración sérica de calcio y su excreción urinaria. El túbulo contorneado distal y conector es donde se produce la regulación fina de la calciuria. A ese nivel se encuentra el canal epitelial de Ca (TRPV5), que es el paso limitante en el transporte transcelular de Ca. La presencia dinámica del canal TRPV5 en la superficie de la célula tubular está mediada por un proceso de reciclado endosómico. Distintos factores intrarrenales intervienen en la fijación del canal de calcio en la membrana aplical, entre ellos la hormona antienvejecimiento klotho y la calicreína tisular (CT). Ambas proteínas son sintetizadas en el túbulo distal y secretadas en el fluido tubular. La calicreína tisular estimula la reabsorción activa de calcio por vía del receptor de bradiquinina tipo 2 que compromete la activación del of TRPV5 por vía de la protein cinasa C. Los ratones deficientes en CT muestran hipercalciuria de origen renal comparable a la pérdida de calcio que se observa en los ratones knockout para el TRPV5. Existe un polimorfismo con pérdida de función del gen de la CT humana denominado R53H (alelo H) que produce una gran disminución de la actividad enzimática. La presencia del alelo H, por lo menos en la población japonesa, parece ser frecuente (24%). Estos individuos tiene una tendencia a excreción más alta de calcio y sodio en orina que se manifiesta más durante la infusión de furosemida. En el futuro habrá que estudiar si la manipulación del sistema calicreína-quinina renal permite corregir la hipercalciuria idiopática con fármacos diferentes a los diuréticos tiazídicos (AU)


Renal tubular calcium reabsorption is one of the principal factors that determine serum calcium concentration and calcium excretion. Calcium excretion is regulated by the distal convoluted tubule and connecting tubule, where the epithelial calcium channel TRPV5 can be found, which limits the rate of transcellular calcium transport. The dynamic presence of the TRPV5 channel on the surface of the tubular cell is mediated by an endosomal recycling process. Different intrarenal factors are involved in calcium channel fixation in the apical membrane, including the anti-ageing hormone klotho and tissue kallikrein (TK). Both proteins are synthesised in the distal tubule and secreted in the tubular fluid. TK stimulates active calcium reabsorption through the bradykinin receptor B2 that compromises TRPV5 activation through the protein kinase C pathway. TK-deficient mice show hypercalciuria of renal origin comparable to that seen in TRPV5 knockout mice. There is a polymorphism with loss of function of the human TK gene R53H (allele H) that causes a marked decrease in enzymatic activity. The presence of the allele H seems to be common at least in the Japanese population (24%). These individuals have a tendency to greater calcium and sodium excretion in urine that is more evident during furosemide infusion. Future studies should analyse if manipulating the renal kallikrein-kinin system can correct idiopathic hypercalciuria with drugs other than thiazide diuretics (AU)


Humans , Kallikrein-Kinin System/physiology , Hypercalciuria/physiopathology , Calcium/metabolism , Renal Reabsorption/physiology
16.
Orphanet J Rare Dis ; 12(1): 19, 2017 01 25.
Article En | MEDLINE | ID: mdl-28122587

The calcium-sensing receptor (CaSR) plays a pivotal role in systemic calcium metabolism by regulating parathyroid hormone secretion and urinary calcium excretion. The diseases caused by an abnormality of the CaSR are genetically determined or are more rarely acquired. The genetic diseases consist of hyper- or hypocalcemia disorders. Hypercalcaemia disorders are related to inactivating mutations of the CASR gene either heterozygous (autosomal dominant familial benign hypercalcaemia, still named hypocalciuric hypercalcaemia syndrome type 1) or homozygous (severe neonatal hyperparathyroidism). The A986S, R990G and Q1011E variants of the CASR gene are associated with higher serum calcium levels than in the general population, hypercalciuria being also associated with the R990G variant. The differential diagnosis consists in the hypocalciuric hypercalcaemia syndrome, types 2 (involving GNA11 gene) and 3 (involving AP2S1 gene); hyperparathyroidism; abnormalities of vitamin D metabolism, involving CYP24A1 and SLC34A1 genes; and reduced GFR. Hypocalcemia disorders, which are more rare, are related to heterozygous activating mutations of the CASR gene (type 1), consisting of autosomal dominant hypocalcemia disorders, sometimes with a presentation of pseudo-Bartter's syndrome. The differential diagnosis consists of the hypercalciuric hypocalcaemia syndrome type 2, involving GNA11 gene and other hypoparathyroidism aetiologies. The acquired diseases are related to the presence of anti-CaSR antibodies, which can cause hyper- or especially hypocalcemia disorders (for instance in APECED syndromes), determined by their functionality. Finally, the role of CaSR in digestive, respiratory, cardiovascular and neoplastic diseases is gradually coming to light, providing new therapeutic possibilities. Two types of CaSR modulators are known: CaSR agonists (or activators, still named calcimimetics) and calcilytic antagonists (or inhibitors of the CasR). CaSR agonists, such as cinacalcet, are indicated in secondary and primary hyperparathyroidism. Calcilytics have no efficacy in osteoporosis, but could be useful in the treatment of hypercalciuric hypocalcaemia syndromes.


Genetic Diseases, Inborn/physiopathology , Receptors, Calcium-Sensing/metabolism , Animals , Calcium/blood , Calcium/metabolism , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Genetic Diseases, Inborn/blood , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/urine , Humans , Hypercalciuria/blood , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Hyperparathyroidism/blood , Hyperparathyroidism/genetics , Hyperparathyroidism/physiopathology , Receptors, Calcium-Sensing/genetics
17.
Physiology (Bethesda) ; 30(4): 317-26, 2015 Jul.
Article En | MEDLINE | ID: mdl-26136545

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.


Calcium/metabolism , Kidney Tubules/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Homeostasis , Humans , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Kidney Tubules/physiopathology , Signal Transduction
18.
Am J Physiol Regul Integr Comp Physiol ; 309(1): R85-92, 2015 Jul 01.
Article En | MEDLINE | ID: mdl-25947170

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.


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
19.
J Bone Miner Res ; 30(11): 1980-93, 2015 Nov.
Article En | MEDLINE | ID: mdl-25967373

Activating mutations of calcium-sensing receptor (CaSR) cause autosomal dominant hypocalcemia (ADH). ADH patients develop hypocalcemia, hyperphosphatemia, and hypercalciuria, similar to the clinical features of hypoparathyroidism. The current treatment of ADH is similar to the other forms of hypoparathyroidism, using active vitamin D3 or parathyroid hormone (PTH). However, these treatments aggravate hypercalciuria and renal calcification. Thus, new therapeutic strategies for ADH are needed. Calcilytics are allosteric antagonists of CaSR, and may be effective for the treatment of ADH caused by activating mutations of CaSR. In order to examine the effect of calcilytic JTT-305/MK-5442 on CaSR harboring activating mutations in the extracellular and transmembrane domains in vitro, we first transfected a mutated CaSR gene into HEK cells. JTT-305/MK-5442 suppressed the hypersensitivity to extracellular Ca(2+) of HEK cells transfected with the CaSR gene with activating mutations in the extracellular and transmembrane domains. We then selected two activating mutations locating in the extracellular (C129S) and transmembrane (A843E) domains, and generated two strains of CaSR knock-in mice to build an ADH mouse model. Both mutant mice mimicked almost all the clinical features of human ADH. JTT-305/MK-5442 treatment in vivo increased urinary cAMP excretion, improved serum and urinary calcium and phosphate levels by stimulating endogenous PTH secretion, and prevented renal calcification. In contrast, PTH(1-34) treatment normalized serum calcium and phosphate but could not reduce hypercalciuria or renal calcification. CaSR knock-in mice exhibited low bone turnover due to the deficiency of PTH, and JTT-305/MK-5442 as well as PTH(1-34) increased bone turnover and bone mineral density (BMD) in these mice. These results demonstrate that calcilytics can reverse almost all the phenotypes of ADH including hypercalciuria and renal calcification, and suggest that calcilytics can become a novel therapeutic agent for ADH.


Benzoates/therapeutic use , Hypercalciuria/drug therapy , Hypercalciuria/genetics , Hypocalcemia/drug therapy , Hypocalcemia/genetics , Hypoparathyroidism/congenital , Mutation/genetics , Propanolamines/therapeutic use , Receptors, Calcium-Sensing/genetics , Animals , Base Sequence , Benzoates/pharmacology , Bone Remodeling/drug effects , Bone and Bones/drug effects , Bone and Bones/pathology , Calcium/metabolism , Disease Models, Animal , Fibroblast Growth Factor-23 , Gene Knock-In Techniques , HEK293 Cells , Humans , Hypercalciuria/pathology , Hypercalciuria/physiopathology , Hypocalcemia/pathology , Hypocalcemia/physiopathology , Hypoparathyroidism/drug therapy , Hypoparathyroidism/genetics , Hypoparathyroidism/pathology , Hypoparathyroidism/physiopathology , Mice , Molecular Sequence Data , Mutant Proteins/metabolism , Organ Size/drug effects , Phenotype , Propanolamines/pharmacology
20.
Ren Fail ; 37(1): 180-3, 2015 Feb.
Article En | MEDLINE | ID: mdl-25366522

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.


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
...