Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Yakugaku Zasshi ; 144(6): 659-674, 2024.
Article in Japanese | MEDLINE | ID: mdl-38825475

ABSTRACT

Serum urate levels are determined by the balance between uric acid production and uric acid excretion capacity from the kidneys and intestinal tract. Dysuricemia, including hyperuricemia and hypouricemia, develops when the balance shifts towards an increase or a decrease in the uric acid pool. Hyperuricemia is mostly a multifactorial genetic disorder involving several disease susceptibility genes and environmental factors. Hypouricemia, on the other hand, is caused by genetic abnormalities. The main genes involved in dysuricemia are xanthine oxidoreductase, an enzyme that produces uric acid, and the urate transporters urate transporter 1/solute carrier family 22 member 12 (URAT1/SLC22A12), glucose transporter 9/solute carrier family 2 member 9 (GLUT9/SLC2A9) and ATP binding cassette subfamily G member 2 (ABCG2). Deficiency of xanthine oxidoreductase results in xanthinuria, a rare disease with marked hypouricemia. Xanthinuria can be due to a single deficiency of xanthine oxidoreductase or in combination with aldehyde oxidase deficiency as well. The latter is caused by a deficiency in molybdenum cofactor sulfurase, which is responsible for adding sulphur atoms to the molybdenum cofactor required for xanthine oxidoreductase and aldehyde oxidase to exert their action. URAT1/SLC22A12 and GLUT9/SLC2A9 are involved in urate reabsorption and their deficiency leads to renal hypouricemia, a condition that is common in Japanese due to URAT1/SLC22A12 deficiency. On the other hand, ABCG2 is involved in the secretion of urate, and many Japanese have single nucleotide polymorphisms that result in its reduced function, leading to hyperuricemia. In particular, severe dysfunction of ABCG2 leads to hyperuricemia with reduced extrarenal excretion.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2 , Glucose Transport Proteins, Facilitative , Hyperuricemia , Neoplasm Proteins , Organic Anion Transporters , Uric Acid , Xanthine Dehydrogenase , Humans , Hyperuricemia/etiology , Hyperuricemia/metabolism , Hyperuricemia/genetics , Uric Acid/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Organic Anion Transporters/metabolism , Organic Anion Transporters/genetics , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transport Proteins, Facilitative/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Xanthine Dehydrogenase/metabolism , Xanthine Dehydrogenase/genetics , Xanthine Dehydrogenase/deficiency , Animals , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/etiology , Renal Tubular Transport, Inborn Errors/metabolism , Urinary Calculi/etiology , Urinary Calculi/metabolism , Urinary Calculi/genetics , Metabolism, Inborn Errors
2.
Am J Physiol Renal Physiol ; 322(1): F14-F26, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34747197

ABSTRACT

The multiligand receptors megalin (Lrp2) and cubilin (Cubn) and their endocytic adaptor protein Dab2 (Dab2) play essential roles in maintaining the integrity of the apical endocytic pathway of proximal tubule (PT) cells and have complex and poorly understood roles in the development of chronic kidney disease. Here, we used RNA-sequencing and CRISPR/Cas9 knockout (KO) technology in a well-differentiated cell culture model to identify PT-specific transcriptional changes that are directly consequent to the loss of megalin, cubilin, or Dab2 expression. KO of Lrp2 had the greatest transcriptional effect, and nearly all genes whose expression was affected in Cubn KO and Dab2 KO cells were also changed in Lrp2 KO cells. Pathway analysis and more granular inspection of the altered gene profiles suggested changes in pathways with immunomodulatory functions that might trigger the pathological changes observed in KO mice and patients with Donnai-Barrow syndrome. In addition, differences in transcription patterns between Lrp2 and Dab2 KO cells suggested the possibility that altered spatial signaling by aberrantly localized receptors contributes to transcriptional changes upon the disruption of PT endocytic function. A reduction in transcripts encoding sodium-glucose cotransporter isoform 2 was confirmed in Lrp2 KO mouse kidney lysates by quantitative PCR analysis. Our results highlight the role of megalin as a master regulator and coordinator of ion transport, metabolism, and endocytosis in the PT. Compared with the studies in animal models, this approach provides a means to identify PT-specific transcriptional changes that are directly consequent to the loss of these target genes.NEW & NOTEWORTHY Megalin and cubilin receptors together with their adaptor protein Dab2 represent major components of the endocytic machinery responsible for efficient uptake of filtered proteins by the proximal tubule (PT). Dab2 and megalin expression have been implicated as both positive and negative modulators of kidney disease. We used RNA sequencing to knock out CRISPR/Cas9 cubilin, megalin, and Dab2 in highly differentiated PT cells to identify PT-specific changes that are directly consequent to knockout of each component.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Gene Knockout Techniques , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Receptors, Cell Surface/metabolism , Transcription, Genetic , Adaptor Proteins, Signal Transducing/genetics , Agenesis of Corpus Callosum/genetics , Agenesis of Corpus Callosum/metabolism , Agenesis of Corpus Callosum/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Databases, Genetic , Gene Regulatory Networks , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/pathology , Hernias, Diaphragmatic, Congenital/genetics , Hernias, Diaphragmatic, Congenital/metabolism , Hernias, Diaphragmatic, Congenital/pathology , Humans , Kidney Tubules, Proximal/pathology , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Mice, Knockout , Monodelphis , Myopia/genetics , Myopia/metabolism , Myopia/pathology , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/pathology , Receptors, Cell Surface/genetics , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/metabolism , Renal Tubular Transport, Inborn Errors/pathology
3.
J Am Soc Nephrol ; 33(2): 326-341, 2022 02.
Article in English | MEDLINE | ID: mdl-34799437

ABSTRACT

BACKGROUND: Hereditary renal hypouricemia type 1 (RHUC1) is caused by URAT1/SLC22A12 dysfunction, resulting in urolithiasis and exercise-induced AKI (EIAKI). However, because there is no useful experimental RHUC1 animal model, the precise pathophysiologic mechanisms underlying EIAKI have yet to be elucidated. We established a high HPRT activity Urat1-Uox double knockout (DKO) mouse as a novel RHUC1 animal model for investigating the cause of EIAKI and the potential therapeutic effect of xanthine oxidoreductase inhibitors (XOIs). METHODS: The novel Urat1-Uox DKO mice were used in a forced swimming test as loading exercise to explore the onset mechanism of EIAKI and evaluate related purine metabolism and renal injury parameters. RESULTS: Urat1-Uox DKO mice had uricosuric effects and elevated levels of plasma creatinine and BUN as renal injury markers, and decreased creatinine clearance observed in a forced swimming test. In addition, Urat1-Uox DKO mice had increased NLRP3 inflammasome activity and downregulated levels of Na+-K+-ATPase protein in the kidney, as Western blot analysis showed. Finally, we demonstrated that topiroxostat and allopurinol, XOIs, improved renal injury and functional parameters of EIAKI. CONCLUSIONS: Urat1-Uox DKO mice are a useful experimental animal model for human RHUC1. The pathogenic mechanism of EIAKI was found to be due to increased levels of IL-1ß via NLRP3 inflammasome signaling and Na+-K+-ATPase dysfunction associated with excessive urinary urate excretion. In addition, XOIs appear to be a promising therapeutic agent for the treatment of EIAKI.


Subject(s)
Acute Kidney Injury/drug therapy , Hypoxanthine Phosphoribosyltransferase/metabolism , Organic Anion Transporters/deficiency , Urate Oxidase/deficiency , Xanthine Dehydrogenase/antagonists & inhibitors , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Allopurinol/pharmacology , Animals , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hypoxanthine Phosphoribosyltransferase/genetics , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nitriles/pharmacology , Organic Anion Transporters/genetics , Physical Exertion , Pyridines/pharmacology , Renal Tubular Transport, Inborn Errors/drug therapy , Renal Tubular Transport, Inborn Errors/etiology , Renal Tubular Transport, Inborn Errors/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Urate Oxidase/genetics , Urinary Calculi/drug therapy , Urinary Calculi/etiology , Urinary Calculi/metabolism
4.
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
5.
Nutrients ; 12(11)2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33114586

ABSTRACT

Hypomagnesemia is very commonly observed in cancer patients, most frequently in association with therapy with cetuximab (CTX), a monoclonal antibody targeting the epithelial growth factor receptor (EGFR). CTX-induced hypomagnesemia has been ascribed to renal magnesium (Mg) wasting. Here, we sought to clarify whether CTX may also influence intestinal Mg absorption and if Mg supplementation may interfere with CTX activity. We used human colon carcinoma CaCo-2 cells as an in vitro model to study the mechanisms underlying Mg transport and CTX activity. Our findings demonstrate that TRPM6 is the key channel that mediates Mg influx in intestinal cells and that EGF stimulates such influx; consequently, CTX downregulates TRPM6-mediated Mg influx by interfering with EGF signaling. Moreover, we show that Mg supplementation does not modify either the CTX IC50 or CTX-dependent inhibition of ERK1/2 phosphorylation. Our results suggest that reduced Mg absorption in the intestine may contribute to the severe hypomagnesemia that occurs in CTX-treated patients, and Mg supplementation may represent a safe and effective nutritional intervention to restore Mg status without impairing the CTX efficacy.


Subject(s)
Cetuximab/adverse effects , Epidermal Growth Factor/metabolism , Intestinal Absorption/drug effects , Magnesium/metabolism , TRPM Cation Channels/metabolism , Caco-2 Cells , Colon/metabolism , Humans , Renal Tubular Transport, Inborn Errors/chemically induced , Renal Tubular Transport, Inborn Errors/metabolism
6.
BMC Nephrol ; 21(1): 282, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32677916

ABSTRACT

BACKGROUND: Renal hypouricemia (RHUC) is a genetic disorder caused by mutations in the SLC22A12 gene, which encodes the major uric acid (UA) transporter, URAT1. The clinical course of related, living donor-derived RHUC in patients undergoing kidney transplantation is poorly understood. Here, we report a case of kidney transplantation from a living relative who had an SLC22A12 mutation. After the transplantation, the recipient's fractional excretion of UA (FEUA) decreased, and chimeric tubular epithelium was observed. CASE PRESENTATION: A 40-year-old man underwent kidney transplantation. His sister was the kidney donor. Three weeks after the transplantation, he had low serum-UA, 148.7 µmol/L, and elevated FEUA, 20.8% (normal: < 10%). The patient's sister had low serum-UA (101.1 µmol/L) and high FEUA (15.8%) before transplant. Suspecting RHUC, we performed next-generation sequencing on a gene panel containing RHUC-associated genes. A heterozygous missense mutation in the SLC22A12 gene was detected in the donor, but not in the recipient. The recipient's serum-UA level increased from 148.7 µmol/L to 231.9 µmol/L 3 months after transplantation and was 226.0 µmol/L 1 year after transplantation. His FEUA decreased from 20.8 to 11.7% 3 months after transplantation and was 12.4% 1 year after transplantation. Fluorescence in situ hybridization of allograft biopsies performed 3 months and 1 year after transplantation showed the presence of Y chromosomes in the tubular epithelial cells, suggesting the recipient's elevated serum-UA levels were owing to a chimeric tubular epithelium. CONCLUSIONS: We reported on a kidney transplant recipient that developed RHUC owing to his donor possessing a heterozygous mutation in the SLC22A12 (URAT1) gene. Despite this mutation, the clinical course was not problematic. Thus, the presence of donor-recipient chimerism in the tubular epithelium might positively affect the clinical course, at least in the short-term.


Subject(s)
Kidney Transplantation , Kidney Tubules/metabolism , Living Donors , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Renal Tubular Transport, Inborn Errors/genetics , Uric Acid/metabolism , Urinary Calculi/genetics , Adult , Chimerism , Female , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Male , Mutation, Missense , Renal Elimination/genetics , Renal Tubular Transport, Inborn Errors/metabolism , Siblings , Urinary Calculi/metabolism
7.
Genes (Basel) ; 11(3)2020 03 05.
Article in English | MEDLINE | ID: mdl-32150856

ABSTRACT

Electrolyte homeostasis is maintained by the kidney through a complex transport function mostly performed by specialized proteins distributed along the renal tubules. Pathogenic variants in the genes encoding these proteins impair this function and have consequences on the whole organism. Establishing a genetic diagnosis in patients with renal tubular dysfunction is a challenging task given the genetic and phenotypic heterogeneity, functional characteristics of the genes involved and the number of yet unknown causes. Part of these difficulties can be overcome by gathering large patient cohorts and applying high-throughput sequencing techniques combined with experimental work to prove functional impact. This approach has led to the identification of a number of genes but also generated controversies about proper interpretation of variants. In this article, we will highlight these challenges and controversies.


Subject(s)
Renal Tubular Transport, Inborn Errors/genetics , Animals , Genetic Heterogeneity , Genetic Testing/methods , Genetic Testing/standards , Humans , Phenotype , Renal Tubular Transport, Inborn Errors/diagnosis , Renal Tubular Transport, Inborn Errors/metabolism
8.
Mol Med Rep ; 20(6): 5118-5124, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31638209

ABSTRACT

Idiopathic renal hypouricemia is a rare hereditary condition. Type 2 renal hyperuricemia (RHUC2) is caused by a mutation in the SLC2A9 gene, which encodes a high­capacity glucose and urate transporter, glucose transporter (GLUT)9. RHUC2 predisposes to exercise­induced acute renal failure (EIARF) and nephrolithiasis, which is caused by a defect in renal tubular urate transport and is characterized by increased clearance of renal uric acid. In the present study a case of a 35­year­old Chinese man with EIARF is reported. The patient had isolated renal hypouricemia, with a serum uric acid level of 21 µmol/l and a fractional excretion of uric acid of 200%. The mutational analysis revealed a homozygous mutation (c.857G>A in exon 8) in the SLC2A9 gene. The patient's family members carried the same mutation, but were heterozygous and clinically asymptomatic. In conclusion, to the best of our knowledge, this is the first report of a RHUC2 patient with a GLUT9 mutation, p.W286X, which may be a pathogenic mutation of RHUC2. Further investigation into the functional role of GLUT9 in this novel SLC2A9 mutation is required.


Subject(s)
Renal Tubular Transport, Inborn Errors/diagnosis , Renal Tubular Transport, Inborn Errors/genetics , Urinary Calculi/diagnosis , Urinary Calculi/genetics , Adult , Alleles , Biomarkers , Biopsy , DNA Mutational Analysis , Glucose Transport Proteins, Facilitative/genetics , Homozygote , Humans , Male , Models, Biological , Mutation , Pedigree , Rare Diseases , Renal Tubular Transport, Inborn Errors/metabolism , Renal Tubular Transport, Inborn Errors/therapy , Sequence Analysis, DNA , Ultrasonography , Urinary Calculi/metabolism , Urinary Calculi/therapy
9.
Pediatr Nephrol ; 34(4): 549-559, 2019 04.
Article in English | MEDLINE | ID: mdl-29275531

ABSTRACT

Renal phosphate handling critically determines plasma phosphate and whole body phosphate levels. Filtered phosphate is mostly reabsorbed by Na+-dependent phosphate transporters located in the brush border membrane of the proximal tubule: NaPi-IIa (SLC34A1), NaPi-IIc (SLC34A3), and Pit-2 (SLC20A2). Here we review new evidence for the role and relevance of these transporters in inherited disorders of renal phosphate handling. The importance of NaPi-IIa and NaPi-IIc for renal phosphate reabsorption and mineral homeostasis has been highlighted by the identification of mutations in these transporters in a subset of patients with infantile idiopathic hypercalcemia and patients with hereditary hypophosphatemic rickets with hypercalciuria. Both diseases are characterized by disturbed calcium homeostasis secondary to elevated 1,25-(OH)2 vitamin D3 as a consequence of hypophosphatemia. In vitro analysis of mutated NaPi-IIa or NaPi-IIc transporters suggests defective trafficking underlying disease in most cases. Monoallelic pathogenic mutations in both SLC34A1 and SLC34A3 appear to be very frequent in the general population and have been associated with kidney stones. Consistent with these findings, results from genome-wide association studies indicate that variants in SLC34A1 are associated with a higher risk to develop kidney stones and chronic kidney disease, but underlying mechanisms have not been addressed to date.


Subject(s)
Kidney Tubules, Proximal/metabolism , Phosphates/metabolism , Renal Reabsorption , Renal Tubular Transport, Inborn Errors/metabolism , Sodium-Phosphate Cotransporter Proteins/metabolism , Animals , Familial Hypophosphatemic Rickets , Fibroblast Growth Factor-23 , Genetic Predisposition to Disease , Heredity , Humans , Mutation , Pedigree , Phenotype , Prognosis , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/physiopathology , Risk Assessment , Risk Factors , Sodium-Phosphate Cotransporter Proteins/genetics , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIc/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIc/metabolism
10.
BMC Med Genet ; 19(1): 142, 2018 08 10.
Article in English | MEDLINE | ID: mdl-30097038

ABSTRACT

BACKGROUND: Renal hypouricemia (RHUC) is a heterogeneous genetic disorder that is characterized by decreased serum uric acid concentration and increased fractional excretion of uric acid. Previous reports have revealed many functional mutations in two urate transporter genes, SLC22A12 and/or SLC2A9, to be the causative genetic factors of this disorder. However, there are still unresolved patients, suggesting the existence of other causal genes or new mutations. Here, we report an RHUC patient with novel compound heterozygous mutations in the SLC22A12 gene. CASE PRESENTATION: A 27-year-old female presenting with recurrent hypouricemia during routine checkups was referred to our hospital. After obtaining the patient's consent, both the patient and her healthy parents were analyzed using whole-exome sequencing (WES) and Sanger sequencing to discover and validate causal mutations, respectively. The prioritization protocol of WES screened out two mutations of c.269G > A/p.R90H and c.1289_1290insGG/p.M430fsX466, which are both located in the SLC22A12 gene, in the patient. Sanger sequencing further confirmed that the patient's heterozygous c.269G > A/p.R90H mutation, which has been reported previously, derived from her mother, and the heterozygous c.1289_1290insGG/p.M430fsX466 mutation, which was found for the first time, derived from her father. p.R90H, which is highly conserved among different species, may decrease the stability of this domain and was considered to be almost damaging in silicon analysis. p.M430fsX466 lacks the last three transmembrane domains, including the tripeptide motif (S/T)XΦ (X = any amino acid and Φ = hydrophobic residue), at the C-terminal, which interact with scaffolding protein PDZK1 and thus will possibly lead to weak functioning of urate transport through the disruption of the "transporter complex" that is formed by URAT1 and PDZK1. CONCLUSIONS: We report a Chinese patient with RHUC, which was caused by compound heterozygous mutations of the SLC22A12 gene, using WES and Sanger sequencing for the first time. Mutation-induced structural instability or malfunction of the urate transporter complex may be the main mechanisms for this hereditary disorder.


Subject(s)
Mutation/genetics , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Renal Tubular Transport, Inborn Errors/genetics , Urinary Calculi/genetics , Adult , Asian People/genetics , Female , Heterozygote , Humans , Male , Renal Tubular Transport, Inborn Errors/metabolism , Uric Acid/metabolism , Urinary Calculi/metabolism
11.
Cell Mol Life Sci ; 75(21): 4021-4040, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29916093

ABSTRACT

Mitochondrial intracrines are extracellular signaling proteins, targeted to the mitochondria. The pathway for mitochondrial targeting of mitochondrial intracrines and actions in the mitochondria remains unknown. Megalin/LRP2 mediates the uptake of vitamins and proteins, and is critical for clearance of amyloid-ß protein from the brain. Megalin mutations underlie the pathogenesis of Donnai-Barrow and Lowe syndromes, characterized by brain defects and kidney dysfunction; megalin was not previously known to reside in the mitochondria. Here, we show megalin is present in the mitochondria and associates with mitochondrial anti-oxidant proteins SIRT3 and stanniocalcin-1 (STC1). Megalin shuttles extracellularly-applied STC1, angiotensin II and TGF-ß to the mitochondria through the retrograde early endosome-to-Golgi transport pathway and Rab32. Megalin knockout in cultured cells impairs glycolytic and respiratory capacities. Thus, megalin is critical for mitochondrial biology; mitochondrial intracrine signaling is a continuum of the retrograde early endosome-to-Golgi-Rab32 pathway and defects in this pathway may underlie disease processes in many systems.


Subject(s)
Amyloid beta-Peptides/genetics , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Mitochondria/genetics , rab GTP-Binding Proteins/genetics , Agenesis of Corpus Callosum/genetics , Agenesis of Corpus Callosum/metabolism , Agenesis of Corpus Callosum/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Cell Membrane/genetics , Glycoproteins/genetics , HEK293 Cells , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/pathology , Hernias, Diaphragmatic, Congenital/genetics , Hernias, Diaphragmatic, Congenital/metabolism , Hernias, Diaphragmatic, Congenital/pathology , Humans , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mice , Mitochondria/metabolism , Myopia/genetics , Myopia/metabolism , Myopia/pathology , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/metabolism , Oculocerebrorenal Syndrome/pathology , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/pathology , RAW 264.7 Cells , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/metabolism , Renal Tubular Transport, Inborn Errors/pathology , Signal Transduction , Sirtuin 3/genetics , Transforming Growth Factor beta/genetics , rab GTP-Binding Proteins/metabolism
12.
Sci Rep ; 7(1): 15623, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29142255

ABSTRACT

In humans, germline mutations in Trpm6 cause autosomal dominant hypomagnesemia with secondary hypocalcemia disorder. Loss of Trpm6 in mice also perturbs cellular magnesium homeostasis but additionally results in early embryonic lethality and neural tube closure defects. To define the mechanisms by which TRPM6 influences neural tube closure, we functionally characterized the role of TRPM6 during early embryogenesis in Xenopus laevis. The expression of Xenopus TRPM6 (XTRPM6) is elevated at the onset of gastrulation and is concentrated in the lateral mesoderm and ectoderm at the neurula stage. Loss of XTRPM6 produced gastrulation and neural tube closure defects. Unlike XTRPM6's close homologue XTRPM7, whose loss interferes with mediolateral intercalation, depletion of XTRPM6 but not XTRPM7 disrupted radial intercalation cell movements. A zinc-influx assay demonstrated that TRPM6 has the potential to constitute functional channels in the absence of TRPM7. The results of our study indicate that XTRPM6 regulates radial intercalation with little or no contribution from XTRPM7 in the region lateral to the neural plate, whereas XTRPM7 is mainly involved in regulating mediolateral intercalation in the medial region of the neural plate. We conclude that both TRPM6 and TRPM7 channels function cooperatively but have distinct and essential roles during neural tube closure.


Subject(s)
Embryonic Development/genetics , Neural Plate/growth & development , Neural Tube/growth & development , TRPM Cation Channels/genetics , Xenopus Proteins/genetics , Animals , Calcium/metabolism , Cell Movement/genetics , Gene Expression Regulation, Developmental , Germ-Line Mutation/genetics , Humans , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/pathology , Hypocalcemia/genetics , Hypocalcemia/metabolism , Hypocalcemia/pathology , Magnesium/metabolism , Nephrocalcinosis/genetics , Nephrocalcinosis/metabolism , Nephrocalcinosis/pathology , Neural Plate/metabolism , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/metabolism , Renal Tubular Transport, Inborn Errors/pathology , Xenopus laevis
13.
Nucleosides Nucleotides Nucleic Acids ; 35(10-12): 543-549, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27906636

ABSTRACT

Renal hypouricemia (RHUC) is a hereditary disease characterized by a low level of plasma urate but with normal urinary urate excretion. RHUC type 1 is caused by mutations of the urate transporter URAT1 gene (SLC22A12). However, the plasma urate levels of URAT1 knockout mice are no different from those of wild-type mice. In the present study, a double knockout mouse, in which the URAT1 and uricase (Uox) genes were deleted (Urat1-Uox-DKO), were used as an experimental animal model of RHUC type 1 to investigate RHUC and excise-induced acute kidney injury (EIAKI). Mice were given a variable content of allopurinol for one week followed by HPLC measurement of urate and creatinine concentrations in spot urine and blood from the tail. The urinary excretion of urate in Urat1-Uox-DKO mice was approximately 25 times higher than those of humans. With allopurinol, the plasma urate levels of Urat1-Uox-DKO mice were lower than those of Uox-KO mice. There were no differences in the urinary urate excretions between Urat1-Uox-DKO and Uox-KO mice administered with 9 mg allopurinol /100 g feed. In the absence of allopurinol, plasma creatinine levels of some Urat1-Uox-DKO mice were higher than those of Uox-KO mice. Consequently, hypouricemia and normouricosuria may indicate that the Urat1-Uox-DKO mouse administered with allopurinol may represent a suitable animal model of RHUC type 1. Urat1-Uox-DKO mice without allopurinol exhibited acute kidney injury, thus providing additional benefit as a potential animal model for EIAKI. Finally, our data indicate that allopurinol appears to provide prophylactic effects for EIAKI.


Subject(s)
Acute Kidney Injury/genetics , Organic Anion Transporters/genetics , Renal Tubular Transport, Inborn Errors/genetics , Urate Oxidase/genetics , Urinary Calculi/genetics , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Allopurinol/pharmacology , Allopurinol/therapeutic use , Animals , Creatinine/blood , Disease Models, Animal , Gout Suppressants/pharmacology , Gout Suppressants/therapeutic use , Male , Mice, Knockout , Organic Anion Transporters/metabolism , Physical Conditioning, Animal , Renal Tubular Transport, Inborn Errors/drug therapy , Renal Tubular Transport, Inborn Errors/metabolism , Urate Oxidase/metabolism , Uric Acid/urine , Urinary Calculi/drug therapy , Urinary Calculi/metabolism
14.
Curr Opin Nephrol Hypertens ; 25(4): 308-13, 2016 07.
Article in English | MEDLINE | ID: mdl-27191348

ABSTRACT

PURPOSE OF REVIEW: The tight junction conductance made of the claudin-based paracellular channel is important in the regulation of calcium and magnesium reabsorption in the kidney. This review describes recent findings of the structure, the function, and the physiologic regulation of claudin-14, claudin-16, and claudin-19 channels that through protein interactions confer calcium and magnesium permeability to the tight junction. RECENT FINDINGS: Mutations in two tight junction genes - claudin-16 and claudin-19 - cause the inherited renal disorder familial hypomagnesemia with hypercalciuria and nephrocalcinosis. A recent genome-wide association study has identified claudin-14 as a major risk gene of hypercalciuric nephrolithiasis. The crystal structure of claudin-19 has recently been resolved allowing the reconstruction of a claudin assembly model from cis-dimers made of claudin-16 and claudin-19 interaction. MicroRNAs have been identified as novel regulators of the claudin-14 gene. The microRNA-claudin-14 operon is directly regulated by the Ca sensing receptor gene in response to hypercalcemia. SUMMARY: The paracellular pathway in the kidney is particularly important for mineral metabolism. Three claudin proteins - claudin-14, claudin-16, and claudin-19 - contribute to the structure and function of this paracellular pathway. Genetic mutations and gene expression changes in these claudins may lead to alteration of the paracellular permeability to calcium and magnesium, ultimately affecting renal mineral metabolism.


Subject(s)
Calcium/metabolism , Claudins/metabolism , Magnesium/metabolism , Tight Junctions/metabolism , Claudins/genetics , Genome-Wide Association Study , Humans , Hypercalciuria/genetics , Hypercalciuria/metabolism , Kidney/metabolism , MicroRNAs/metabolism , Nephrocalcinosis/genetics , Nephrocalcinosis/metabolism , Nephrolithiasis/genetics , Nephrolithiasis/metabolism , Receptors, Calcium-Sensing/metabolism , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/metabolism
15.
Curr Opin Nephrol Hypertens ; 25(4): 301-7, 2016 07.
Article in English | MEDLINE | ID: mdl-27219040

ABSTRACT

PURPOSE OF REVIEW: Identification of the mechanisms of magnesium absorption and reabsorption has markedly enhanced our understanding of the causes of hypomagnesemia. RECENT FINDINGS: New gastrointestinal and renal causes of hypomagnesemia have been recently documented. SUMMARY: The recognition of new mechanisms and causes of magnesium absorption and reabsorption should enhance the ability to monitor patients at risk for hypomagnesemia and improve our ability to mitigate the serious symptoms associated with this disorder.


Subject(s)
Intestinal Elimination , Magnesium/metabolism , Renal Elimination , Water-Electrolyte Imbalance/metabolism , Bartter Syndrome/complications , Bartter Syndrome/metabolism , Gitelman Syndrome/complications , Gitelman Syndrome/metabolism , Humans , Hypercalciuria/complications , Hypercalciuria/metabolism , Hypocalcemia/complications , Hypocalcemia/metabolism , Hypoparathyroidism/complications , Hypoparathyroidism/congenital , Hypoparathyroidism/metabolism , Intestinal Mucosa/metabolism , Magnesium Deficiency/complications , Magnesium Deficiency/congenital , Magnesium Deficiency/metabolism , Nephrocalcinosis/complications , Nephrocalcinosis/metabolism , Nephrons/metabolism , Pancreatitis/complications , Pancreatitis/metabolism , Proton Pump Inhibitors/adverse effects , Renal Tubular Transport, Inborn Errors/complications , Renal Tubular Transport, Inborn Errors/metabolism , Sodium Potassium Chloride Symporter Inhibitors/adverse effects , Water-Electrolyte Imbalance/chemically induced , Water-Electrolyte Imbalance/etiology
16.
Pediatr Nephrol ; 31(3): 407-18, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26178649

ABSTRACT

At least three renal tubular segments are involved in the pathophysiology of salt-losing tubulopathies (SLTs). Whether the pathogenesis starts either in the thick ascending limb of the loop of Henle (TAL) or in the distal convoluted tubule (DCT), it is the function of the downstream-localized aldosterone sensitive distal tubule (ASDT) to contribute to the adaptation process. In isolated TAL defects (loop disorders) ASDT adaptation is supported by upregulation of DCT, whereas in DCT disorders the ASDT is complemented by upregulation of TAL function. This upregulation has a major impact on the clinical presentation of SLT patients. Taking into account both the symptoms and signs of primary tubular defect and of the secondary reactions of adaptation, a clinical diagnosis can be made that eventually leads to an appropriate therapy. In addition to salt wasting, as occurs in all SLTs, characteristic features of loop disorders are hypo- or isosthenuric polyuria and hypercalciuria, whereas characteristics of DCT disorders are hypokalemia and (symptomatic) hypomagnesemia. In both SLT categories, replacement of urinary losses is the primary goal of treatment. In loop disorders COX inhibitors are also recommended to mitigate polyuria, and in DCT disorders magnesium supplementation is essential for effective treatment. Of note, the combination of a salt- and potassium-rich diet together with an adequate fluid intake is always the basis of long-term treatment in all SLTs.


Subject(s)
Kidney Tubules, Distal/physiopathology , Renal Tubular Transport, Inborn Errors/physiopathology , Water-Electrolyte Balance , Adaptation, Physiological , Animals , Calcium/metabolism , Humans , Hyperaldosteronism/etiology , Hyperaldosteronism/physiopathology , Kidney Tubules, Distal/drug effects , Kidney Tubules, Distal/metabolism , Magnesium/metabolism , Renal Agents/therapeutic use , Renal Reabsorption , Renal Tubular Transport, Inborn Errors/complications , Renal Tubular Transport, Inborn Errors/drug therapy , Renal Tubular Transport, Inborn Errors/metabolism , Sodium Chloride/metabolism , Water/metabolism , Water-Electrolyte Balance/drug effects
18.
Nephrol Dial Transplant ; 30(6): 952-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25765846

ABSTRACT

BACKGROUND: Magnesium (Mg(2+)) is an essential ion for cell growth, neuroplasticity and muscle contraction. Blood Mg(2+) levels <0.7 mmol/L may cause a heterogeneous clinical phenotype, including muscle cramps and epilepsy and disturbances in K(+) and Ca(2+) homeostasis. Over the last decade, the genetic origin of several familial forms of hypomagnesaemia has been found. In 2000, mutations in FXYD2, encoding the γ-subunit of the Na(+)-K(+)-ATPase, were identified to cause isolated dominant hypomagnesaemia (IDH) in a large Dutch family suffering from hypomagnesaemia, hypocalciuria and chondrocalcinosis. However, no additional patients have been identified since then. METHODS: Here, two families with hypomagnesaemia and hypocalciuria were screened for mutations in the FXYD2 gene. Moreover, the patients were clinically and genetically characterized. RESULTS: We report a p.Gly41Arg FXYD2 mutation in two families with hypomagnesaemia and hypocalciuria. Interestingly, this is the same mutation as was described in the original study. As in the initial family, several patients suffered from muscle cramps, chondrocalcinosis and epilepsy. Haplotype analysis revealed an overlapping haplotype in all families, suggesting a founder effect. CONCLUSIONS: The recurrent p.Gly41Arg FXYD2 mutation in two new families with IDH confirms that FXYD2 mutation causes hypomagnesaemia. Until now, no other FXYD2 mutations have been reported which could indicate that other FXYD2 mutations will not cause hypomagnesaemia or are embryonically lethal.


Subject(s)
Hypercalciuria/genetics , Magnesium/blood , Mutation/genetics , Nephrocalcinosis/genetics , Renal Tubular Transport, Inborn Errors/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Adult , Female , Genes, Dominant , Homeostasis/genetics , Humans , Hypercalciuria/metabolism , Male , Nephrocalcinosis/metabolism , Pedigree , Renal Tubular Transport, Inborn Errors/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
19.
Eur J Med Genet ; 58(5): 293-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25682901

ABSTRACT

Donnai-Barrow syndrome (DBS; MIM 222448) is characterized by typical craniofacial anomalies (major hypertelorism with bulging eyes), high grade myopia, deafness and low molecular weight proteinuria. The disorder results from mutations in the low density lipoprotein receptor-related protein 2 gene LRP2 that maps to chromosome 2q31.1. LRP2 encodes megalin, a multi-ligand endocytic receptor. Herein, we describe the clinical presentation of 4 patients from 2 unrelated Saudi families. Two novel LRP2 mutations, a homozygous nonsense mutation (c.4968C>G; p.Tyr1656*) and a missense mutation (c.12062G>A; p.Cys4021Tyr), were detected in the first and second family respectively. Interestingly, intrafamilial phenotypic variability was observed in one family, while DBS features were atypical in the second family. Differential diagnosis of DBS includes several syndromes associating hypertelorism with high grade myopia, and several syndromal forms of CDH, which are briefly summarized in this study.


Subject(s)
Agenesis of Corpus Callosum/genetics , Hearing Loss, Sensorineural/genetics , Hernias, Diaphragmatic, Congenital/genetics , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Myopia/genetics , Proteinuria/genetics , Renal Tubular Transport, Inborn Errors/genetics , Adolescent , Agenesis of Corpus Callosum/metabolism , Child , Child, Preschool , Codon, Nonsense , DNA Mutational Analysis , Female , Gene Expression , Hearing Loss, Sensorineural/metabolism , Hernias, Diaphragmatic, Congenital/metabolism , Humans , Infant , Infant, Newborn , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mutation, Missense , Myopia/metabolism , Proteinuria/metabolism , Renal Tubular Transport, Inborn Errors/metabolism , Young Adult
20.
Compr Physiol ; 5(1): 45-98, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25589264

ABSTRACT

The distal convoluted tubule (DCT) is a short nephron segment, interposed between the macula densa and collecting duct. Even though it is short, it plays a key role in regulating extracellular fluid volume and electrolyte homeostasis. DCT cells are rich in mitochondria, and possess the highest density of Na+/K+-ATPase along the nephron, where it is expressed on the highly amplified basolateral membranes. DCT cells are largely water impermeable, and reabsorb sodium and chloride across the apical membrane via electroneurtral pathways. Prominent among this is the thiazide-sensitive sodium chloride cotransporter, target of widely used diuretic drugs. These cells also play a key role in magnesium reabsorption, which occurs predominantly, via a transient receptor potential channel (TRPM6). Human genetic diseases in which DCT function is perturbed have provided critical insights into the physiological role of the DCT, and how transport is regulated. These include Familial Hyperkalemic Hypertension, the salt-wasting diseases Gitelman syndrome and EAST syndrome, and hereditary hypomagnesemias. The DCT is also established as an important target for the hormones angiotensin II and aldosterone; it also appears to respond to sympathetic-nerve stimulation and changes in plasma potassium. Here, we discuss what is currently known about DCT physiology. Early studies that determined transport rates of ions by the DCT are described, as are the channels and transporters expressed along the DCT with the advent of molecular cloning. Regulation of expression and activity of these channels and transporters is also described; particular emphasis is placed on the contribution of genetic forms of DCT dysregulation to our understanding.


Subject(s)
Kidney Tubules, Distal/physiology , Animals , Biological Transport/physiology , Electrophysiological Phenomena/physiology , Humans , Kidney Tubules, Distal/anatomy & histology , Kidney Tubules, Distal/metabolism , Magnesium/metabolism , Potassium/metabolism , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/metabolism , Renal Tubular Transport, Inborn Errors/physiopathology , Sodium Chloride/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...