Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
1.
Cell Signal ; 112: 110887, 2023 12.
Article in English | MEDLINE | ID: mdl-37717713

ABSTRACT

Sirtuin1 (Sirt1) activation significantly attenuated calcium oxalate (CaOx) crystal deposition and renal inflammatory injury by regulating renal immune microenvironment. Here, to elucidate the molecular mechanism underlying the therapeutic effects of Sirt1 on macrophage related inflammation and tubular epithelial cells (TECs) necrosis, we constructed a macrophage and CaOx monohydrate (COM)-stimulated tubular cell co-culture system to mimic immune microenvironment in kidney and established a mouse model of CaOx nephrocalcinosis in wild-type and myeloid-specific Sirt1 knockout mice. Target prediction analyses of Gene Expression Omnibus Datasets showed that only miR-34b-5p is regulated by lipopolysaccharides and upregulated by SRT1720 and targets the TLR4 3'-untranslated region. In vitro, SRT1720 suppressed TLR4 expression and M1 macrophage polarization and decreased reactive oxygen species (ROS) production and mitochondrial damage in COM-stimulated TECs by targeting miR-34b-5p. Mechanically, Sirt1 promoted miR-34b-5p expression by suppressing the tri-methylation of H3K27, which directly bound to the miR-34b-5p promoter and abolished the miR-34b-5p transcription. Furthermore, loss of Sirt1 aggravated CaOx nephrocalcinosis-induced inflammatory and oxidative kidney injury, while AgomiR-34b reversed these effects. Therefore, our data suggested that Sirt1 inhibited TLR4 signaling and M1 macrophage polarization and decreased inflammatory and oxidative injury of TECs in vitro and in vivo.


Subject(s)
MicroRNAs , Nephrocalcinosis , Mice , Animals , Calcium Oxalate/metabolism , Calcium Oxalate/pharmacology , Nephrocalcinosis/metabolism , Sirtuin 1/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Kidney/metabolism , Macrophages/metabolism
2.
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
3.
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
4.
Antioxid Redox Signal ; 38(10-12): 731-746, 2023 04.
Article in English | MEDLINE | ID: mdl-36242511

ABSTRACT

Aims: Calcium oxalate (CaOx) crystal deposition induces damage to the renal tubular epithelium, increases epithelial adhesion, and contributes to CaOx nephrocalcinosis. The long noncoding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) is thought to be involved in this process. In this study, we aimed to investigate the mechanism by which NEAT1 regulates renal tubular epithelium in response to inflammatory and oxidative injury triggered by CaOx crystals. Results: As CaOx crystals were deposited in mouse kidney tissue, the expression of NEAT1 was significantly elevated and positively correlated with interferon regulatory factor 1 (IRF1), Toll-like receptor 4 (TLR4), and NF-κB. NEAT1 targets and inhibits miR-130a-3p as a competitor to endogenous RNA. miR-130 binds to and exerts inhibitory effects on the 3'-untranslated region of IRF1. After transfected with silence-NEAT1, IRF1, TLR4, and NF-κB were also variously inhibited, and oxidative damage in renal calcinosis was subsequently attenuated. When we simultaneously inhibited NEAT1 and miR-130, renal tubular injury was exacerbated. Innovation and Conclusion: We found that the lncRNA NEAT1 can enhance IRF1 signaling through targeted repression of miR-130a-3p and activate TLR4/NF-κB pathways to promote oxidative damage during CaOx crystal deposition. This provides an explanation for the tubular epithelial damage caused by CaOx crystals and offers new ideas and drug targets for the prevention and treatment of CaOx nephrocalcinosis. Antioxid. Redox Signal. 38, 731-746.


Subject(s)
Calcinosis , MicroRNAs , Nephrocalcinosis , RNA, Long Noncoding , Mice , Animals , Calcium Oxalate/chemistry , Calcium Oxalate/metabolism , Calcium Oxalate/pharmacology , Nephrocalcinosis/metabolism , Toll-Like Receptor 4/metabolism , RNA, Long Noncoding/genetics , NF-kappa B/metabolism , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/pharmacology , Kidney/metabolism , Oxidative Stress , MicroRNAs/genetics , Oxidation-Reduction
5.
Oxid Med Cell Longev ; 2022: 4345037, 2022.
Article in English | MEDLINE | ID: mdl-35251472

ABSTRACT

Intrarenal calcium oxalate (CaOx) crystals induce renal tubular epithelial cell (TEC) inflammatory and oxidative injury. This study is aimed at exploring potential therapeutic lipid components in kidney stones because lipids are involved in the development of several diseases and indicate the risk of kidney stones. Serum specimens were collected from 35 kidney stone patients and 35 normal controls. The lipid components in serum were measured, and differences were analyzed. The documented biological importance was comprehensively reviewed to identify lipids that differed significantly between the two groups to find potential agents associated with kidney stones. CaOx nephrocalcinosis mouse model was established to examine the therapeutic effects of specific lipids on CaOx deposition and CaOx-induced oxidative renal injury. Several lipids with significantly different levels were present in the serum of patients with stones and normal controls. Resolvin D1 (RvD1) (4.93-fold change, P < 0.001) and protectin D1 (PD1) (5.06-fold change, P < 0.001) were significantly decreased in the serum of patients with kidney stones, and an integrative review suggested that these factors might be associated with inflammatory responses, which is a crucial mechanism associated with stone damage. The administration of RvD1 and PD1 significantly inhibited kidney CaOx deposition and suppressed CaOx-induced renal tubular cell inflammatory injury and necrosis in a CaOx nephrocalcinosis mouse model. Furthermore, RvD1 and PD1 facilitated the expression of the oxidative indicator superoxide dismutase 2 (SOD2), inhibited NADPH oxidase 2 (NOX2) expression, and diminished intracellular reactive oxygen species (ROS) levels. This study preliminarily elucidated the role of lipids in kidney stones. The inhibitory effects of RvD1 and PD1 on oxidative damage induced by CaOx deposition provide a promising perspective for kidney stone treatment strategies.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/blood , Kidney Calculi/blood , Nephrocalcinosis/drug therapy , Signal Transduction/drug effects , Adult , Aged , Animals , Calcium Oxalate/metabolism , Case-Control Studies , Disease Models, Animal , Female , Glyoxylates/adverse effects , Humans , Kidney Tubules/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Nephrocalcinosis/chemically induced , Nephrocalcinosis/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
6.
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
7.
Oxid Med Cell Longev ; 2021: 1676152, 2021.
Article in English | MEDLINE | ID: mdl-34512861

ABSTRACT

The roles of the lncRNA X inactive specific transcript (XIST) in many diseases, including cancers and inflammatory sickness, have been previously elucidated. However, renal calculus remained poorly understood. In this study, we revealed the potential effects of XIST on kidney stones that were exerted via inflammatory response and oxidative stress mechanisms. We established a glyoxylate-induced calcium oxalate (CaOx) stone mouse model and exposed HK-2 cells to calcium oxalate monohydrate (COM). The interactions among XIST, miR-223-3p, and NOD-like receptor protein 3 (NLRP3) and their respective effects were determined by RNAs and protein expression, luciferase activity, and immunohistochemistry (IHC) assays. Cell necrosis, reactive oxygen species (ROS) generation, and inflammatory responses were detected after silencing XIST, activating and inhibiting miR-223-3p, and both knocking down XIST and activating miR-223-3p in vitro and in vivo. The XIST, NLRP3, caspase-1, and IL-1ß levels were notably increased in kidney samples from glyoxylate-induced CaOx stone model mice. XIST knockdown significantly suppressed the inflammatory damage and ROS production and further attenuated oxalate crystal deposition. miRNA-223-3p mimics also exerted the same effects. Moreover, we verified the interactions among XIST, miRNA-223-3p and NLRP3, and the subsequent effects. Our results suggest that the lncRNA XIST participates in the formation and progression of renal calculus by interacting with miR-223-3p and the NLRP3/Caspase-1/IL-1ß pathway to mediate the inflammatory response and ROS production.


Subject(s)
MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephrocalcinosis/drug therapy , Oxidative Stress/drug effects , RNA, Long Noncoding/antagonists & inhibitors , Animals , Calcium Oxalate/administration & dosage , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Nephrocalcinosis/metabolism , Nephrocalcinosis/pathology , Transfection
8.
Am J Physiol Renal Physiol ; 320(3): F475-F484, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33491567

ABSTRACT

Primary hyperoxaluria type 1 (PH1) is a severe inherited disorder caused by a genetic defect in alanine-glyoxylate aminotransferase (AGXT), which results in recurrent urolithiasis and renal failure. Animal models that precisely reflect human PH1 phenotypes are lacking. We aimed to develop a novel PH1 rat model and study the mechanisms involved in PH1 deterioration. One cell stage Sprague-Dawley embryos were injected with the CRISPR/Cas9 system to introduce a Q84X mutation in Agxt. Liver tissues were harvested to determine Agxt expression. Urine oxalate, crystals, and electrolyte levels in AgxtQ84X and wild-type (WT) littermates were evaluated. Kidney tissues were used for Pizzolato staining and kidney injury evaluation. Data showed that Agxt mRNA and protein were absent in AgxtQ84X rats. At 4 and 24 wk, AgxtQ84X rats displayed 2.1- and 2.9-fold higher urinary oxalate levels, respectively, compared with WT littermates. As a result, calcium oxalate (CaOx) crystals in urine were revealed in all AgxtQ84X rats but in none of the WT rats. We also observed bladder stones in 36.4% of AgxtQ84X rats, of which 44.4% had renal CaOx deposition. Moreover, the elevated serum urea and creatinine levels indicated the impaired renal function in AgxtQ84X rats. Further investigation revealed significantly increased expression of inflammation-, necroptosis-, and fibrosis-related genes in the kidneys of AgxtQ84X rats with spontaneous renal CaOx deposition, indicating that these pathways are involved in PH1 deterioration. Collectively, these results suggest that this rat model has broad applicability in mechanistic studies and innovative therapeutics development for PH1 and other kidney stone diseases.NEW & NOTEWORTHY Primary hyperoxaluria type 1 is a severe inherited disorder that results in recurrent urolithiasis and renal failure. We generated an alanine-glyoxylate aminotransferase (Agxt)Q84X nonsense mutant rat model that displayed an early onset of hyperoxaluria, spontaneous renal CaOx precipitation, bladder stone, and kidney injuries. Our results suggest an interaction of renal CaOx crystals with the activation of inflammation-, fibrosis-, and necroptosis-related pathways. In all, the AgxtQ84X rat strain has broad applicability in mechanistic studies and the development of innovative therapeutics.


Subject(s)
Hyperoxaluria/metabolism , Kidney/metabolism , Nephrocalcinosis/metabolism , Transaminases/genetics , Animals , Calcium Oxalate/metabolism , Hyperoxaluria/genetics , Kidney Calculi/blood , Mutation/genetics , Nephrocalcinosis/genetics , Oxalates/metabolism , Rats , Renal Insufficiency/genetics , Transaminases/metabolism
9.
Endocr J ; 68(1): 111-117, 2021 Jan 28.
Article in English | MEDLINE | ID: mdl-32879144

ABSTRACT

SHORT syndrome is a rare developmental disorder frequently associated with growth failure and insulin resistant diabetes mellitus (IRDM). Since GH has a diabetogenic effect, GH therapy has been regarded as a contraindication. We observed a Brazilian girl with SHORT syndrome who received GH therapy from 4 6/12 years of age for SGA short stature. GH dosage was increased from 0.23 to 0.36 mg/kg/week, but statural response to GH therapy remained poor. Her blood HbA1c level, though it remained 5.5-6.0% in childhood, began to elevate with puberty and increased to 9.2% at 10 6/12 years of age, despite the discontinuation of GH therapy at 9 11/12 years of age. Laboratory studies indicated antibody-negative IRDM. She was treated with metformin and canagliflozin (a sodium glucose co-transporter 2 (SGLT2) inhibitor), which ameliorated overt diurnal hyperglycemia and mild nocturnal hypoglycemia and reduced her blood HbA1c around 7%. Whole exome sequencing revealed a de novo heterozygous pathogenic variant (c.1945C>T:p.(Arg649Trp)) in PIK3R1 known as the sole causative gene for SHORT syndrome. Subsequent literature review for patients with molecularly confirmed SHORT syndrome revealed the development of IRDM in 10 of 15 GH-untreated patients aged ≥12 years but in none of three GH-treated and six GH-untreated patients aged ≤10 years. These findings imply a critical role of pubertal development and/or advanced age rather than GH therapy in the development of IRDM, and a usefulness of SGLT2 inhibitor in the treatment of IRDM.


Subject(s)
Diabetes Mellitus/diagnosis , Growth Disorders/complications , Hypercalcemia/complications , Insulin Resistance/physiology , Metabolic Diseases/complications , Nephrocalcinosis/complications , Brazil , Canagliflozin/administration & dosage , Child , Diabetes Complications/diagnosis , Diabetes Complications/drug therapy , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Drug Therapy, Combination , Female , Growth Disorders/diagnosis , Growth Disorders/drug therapy , Growth Disorders/metabolism , Human Growth Hormone/administration & dosage , Humans , Hypercalcemia/diagnosis , Hypercalcemia/drug therapy , Hypercalcemia/metabolism , Metabolic Diseases/diagnosis , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Metformin/administration & dosage , Nephrocalcinosis/diagnosis , Nephrocalcinosis/drug therapy , Nephrocalcinosis/metabolism , Puberty/drug effects , Puberty/metabolism , Sodium-Glucose Transporter 2 Inhibitors/administration & dosage
10.
Hematology Am Soc Hematol Educ Program ; 2020(1): 115-122, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33275732

ABSTRACT

The short telomere syndromes encompass a spectrum of clinical manifestations that present from infancy to late adulthood. They are caused by mutations in telomerase and other telomere maintenance genes and have a predominantly degenerative phenotype characterized by organ failure across multiple systems. They are collectively one of the most common inherited bone marrow failure syndromes; however, their most prevalent presentations are extrahematopoietic. This review focuses on these common nonhematologic complications, including pulmonary fibrosis, liver pathology, and immunodeficiency. The short telomere syndrome diagnosis informs clinical care, especially in guiding diagnostic evaluations as well as in the solid organ transplant setting. Early recognition allows an individualized approach to screening and management. This review illustrates a myriad of extrahematopoietic presentations of short telomere syndromes and how they impact clinical decisions.


Subject(s)
Growth Disorders , Hypercalcemia , Metabolic Diseases , Nephrocalcinosis , Telomere , Animals , Female , Growth Disorders/diagnosis , Growth Disorders/genetics , Growth Disorders/metabolism , Growth Disorders/pathology , Humans , Hypercalcemia/diagnosis , Hypercalcemia/genetics , Hypercalcemia/metabolism , Hypercalcemia/pathology , Metabolic Diseases/diagnosis , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Mice , Middle Aged , Nephrocalcinosis/diagnosis , Nephrocalcinosis/genetics , Nephrocalcinosis/metabolism , Nephrocalcinosis/pathology , Telomere/genetics , Telomere/metabolism , Telomere/pathology
11.
Regul Toxicol Pharmacol ; 117: 104776, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32871170

ABSTRACT

When extrapolating data from animal toxicological studies a default factor (dUF) of 100 is applied to derive a heath based guidance value. The UF takes into account the interspecies differences (ID) and the intraspecies variability (IV). When re-evaluating the safety of phosphates used as food additives nephrocalcinosis was identified as the critical endpoint. The underlying mechanism for nephrocalcinosis was attributed to the precipitation of calcium phosphate in the kidney, depending on its solubility, irrespective of the species and the population. Based on the mechanism, the volume of primary urine, for which the glomerular filtration rate (GFR) was used as a proxy, was considered to be the only parameter relevant for ID and IV. Median value of GFR in rats was 4.0 ml/min/kg bw. In humans it was 1.6 ml/min/kg bw in healthy adults and 0.9 in elderly. These values were calculated from the distribution of the GFR data from 8 studies in rats (n = 191), 16 studies in adults (n = 1540) and 5 studies in elderly (n = 2608). Multiplying the distribution of the ratio rat/healthy humans (ID) with the distribution of the ratio healthy humans/elderly human (IV) resulted in a phosphate specific factor of 4.5 (3.3-6.7) (median; 25th - 75th percentile).


Subject(s)
Calcium Phosphates/toxicity , Glomerular Filtration Rate/drug effects , Kidney/drug effects , Nephrocalcinosis/chemically induced , Animals , Calcium Phosphates/metabolism , Glomerular Filtration Rate/physiology , Humans , Kidney/metabolism , Nephrocalcinosis/metabolism , Nephrocalcinosis/physiopathology , Rats , Risk Assessment , Species Specificity
12.
Am J Physiol Renal Physiol ; 319(2): F202-F214, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32628541

ABSTRACT

Kidney stone disease is a crystal concretion formed in the kidneys that has been associated with an increased risk of chronic kidney disease. MicroRNAs are functionally involved in kidney injury. Data mining using a microRNA array database suggested that miR-21 may be associated with calcium oxalate monohydrate (COM)-induced renal tubular cell injury. Here, we confirmed that COM exposure significantly upregulated miR-21 expression, inhibited proliferation, promoted apoptosis, and caused lipid accumulation in an immortalized renal tubular cell line (HK-2). Moreover, inhibition of miR-21 enhanced proliferation and decreased apoptosis and lipid accumulation in HK-2 cells upon COM exposure. In a glyoxylate-induced mouse model of renal calcium oxalate deposition, increased miR-21 expression, lipid accumulation, and kidney injury were also observed. In silico analysis and subsequent experimental validation confirmed the peroxisome proliferator-activated receptor (PPAR)-α gene (PPARA) a key gene in fatty acid oxidation, as a direct miR-21 target. Suppression of miR-21 by miRNA antagomiR or activation of PPAR-α by its selective agonist fenofibrate significantly reduced renal lipid accumulation and protected against renal injury in vivo. In addition, miR-21 was significantly increased in urine samples from patients with calcium oxalate renal stones compared with healthy volunteers. In situ hybridization of biopsy samples from patients with nephrocalcinosis revealed that miR-21 was also significantly upregulated compared with normal kidney tissues from patients with renal cell carcinoma who underwent radical nephrectomy. These results suggested that miR-21 promoted calcium oxalate-induced renal tubular cell injury by targeting PPARA, indicating that miR-21 could be a potential therapeutic target and biomarker for nephrolithiasis.


Subject(s)
Calcium Oxalate/pharmacology , Kidney/injuries , MicroRNAs/pharmacology , PPAR alpha/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers/metabolism , Calcium Oxalate/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Kidney/metabolism , Kidney Calculi/pathology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , MicroRNAs/genetics , Nephrocalcinosis/metabolism , Signal Transduction/drug effects
13.
Mol Metab ; 40: 101020, 2020 10.
Article in English | MEDLINE | ID: mdl-32439336

ABSTRACT

OBJECTIVE: Insulin signalling via phosphoinositide 3-kinase (PI3K) requires PIK3R1-encoded regulatory subunits. C-terminal PIK3R1 mutations cause SHORT syndrome, as well as lipodystrophy and insulin resistance (IR), surprisingly without fatty liver or metabolic dyslipidaemia. We sought to investigate this discordance. METHODS: The human pathogenic Pik3r1 Y657∗ mutation was knocked into mice by homologous recombination. Growth, body composition, bioenergetic and metabolic profiles were investigated on chow and high-fat diet (HFD). We examined adipose and liver histology, and assessed liver responses to fasting and refeeding transcriptomically. RESULTS: Like humans with SHORT syndrome, Pik3r1WT/Y657∗ mice were small with severe IR, and adipose expansion on HFD was markedly reduced. Also as in humans, plasma lipid concentrations were low, and insulin-stimulated hepatic lipogenesis was not increased despite hyperinsulinemia. At odds with lipodystrophy, however, no adipocyte hypertrophy nor adipose inflammation was found. Liver lipogenic gene expression was not significantly altered, and unbiased transcriptomics showed only minor changes, including evidence of reduced endoplasmic reticulum stress in the fed state and diminished Rictor-dependent transcription on fasting. Increased energy expenditure, which was not explained by hyperglycaemia nor intestinal malabsorption, provided an alternative explanation for the uncoupling of IR from dyslipidaemia. CONCLUSIONS: Pik3r1 dysfunction in mice phenocopies the IR and reduced adiposity without lipotoxicity of human SHORT syndrome. Decreased adiposity may not reflect bona fide lipodystrophy, but rather, increased energy expenditure, and we suggest that further study of brown adipose tissue in both humans and mice is warranted.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/genetics , Growth Disorders/metabolism , Hypercalcemia/metabolism , Insulin Resistance/genetics , Metabolic Diseases/metabolism , Nephrocalcinosis/metabolism , Adipose Tissue, Brown/metabolism , Adiposity , Animals , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Diet, High-Fat , Dyslipidemias/genetics , Energy Metabolism/genetics , Fatty Liver/metabolism , Growth Disorders/genetics , Hypercalcemia/genetics , Inflammation/metabolism , Insulin/metabolism , Lipogenesis , Liver/metabolism , Male , Metabolic Diseases/genetics , Mice , Mice, Inbred C57BL , Nephrocalcinosis/genetics , Obesity/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism
14.
Sci Rep ; 10(1): 4864, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32184468

ABSTRACT

Excessive phosphorus intake adversely affects bone and mineral metabolism. Estrogen is one of the factors affecting fibroblast growth factor 23 (FGF23), a phosphorus-regulating hormone. However, the interaction between excess phosphorus and estrogen status has not been fully elucidated. This study investigated the involvement of estrogen in the effects of high phosphorus intake on bone metabolism and ectopic calcification in ovariectomized (OVX) rats. The interaction between high phosphorus diet and OVX was not observed in bone mineral density and aortic calcium. In contrast, high phosphorus intake markedly increased renal calcium concentration in sham rats, whereas the effect was attenuated in OVX rats, which was reversed by a selective estrogen-receptor modulator treatment. A strong positive correlation between renal calcium and serum FGF23 was observed. In addition, fibroblast growth factor receptor 1 (FGFR1: a predominant receptor of FGF23) inhibitor treatment partially decreased renal calcium concentrations in rats with high phosphorus intake. In conclusion, the effect of high phosphorus intake on bone metabolism and aortic calcification did not depend on the estrogen status; in contrast, high phosphorus intake synergistically induced nephrocalcinosis in the presence of estrogenic action on the bone. Furthermore, FGF23 was involved in the nephrocalcinosis induced by high phosphorus intake partially through FGFR1 signaling.


Subject(s)
Estrogens/metabolism , Fibroblast Growth Factors/blood , Nephrocalcinosis/metabolism , Phosphorus/adverse effects , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Aorta/metabolism , Bone Density/drug effects , Calcium/metabolism , Disease Models, Animal , Female , Fibroblast Growth Factors/drug effects , Nephrocalcinosis/blood , Nephrocalcinosis/chemically induced , Ovariectomy/adverse effects , Pyrimidines/pharmacology , Raloxifene Hydrochloride/pharmacology , Rats
15.
Genes (Basel) ; 11(3)2020 03 10.
Article in English | MEDLINE | ID: mdl-32164158

ABSTRACT

Claudins are integral proteins expressed at the tight junctions of epithelial and endothelial cells. In the mammalian kidney, every tubular segment express a specific set of claudins that give to that segment unique properties regarding permeability and selectivity of the paracellular pathway. So far, 3 claudins (10b, 16 and 19) have been causally traced to rare human syndromes: variants of CLDN10b cause HELIX syndrome and variants of CLDN16 or CLDN19 cause familial hypomagnesemia with hypercalciuria and nephrocalcinosis. The review summarizes our current knowledge on the physiology of mammalian tight junctions and paracellular ion transport, as well as on the role of the 3 above-mentioned claudins in health and disease. Claudin 14, although not having been causally linked to any rare renal disease, is also considered, because available evidence suggests that it may interact with claudin 16. Some single-nucleotide polymorphisms of CLDN14 are associated with urinary calcium excretion and/or kidney stones. For each claudin considered, the pattern of expression, the function and the human syndrome caused by pathogenic variants are described.


Subject(s)
Claudins/metabolism , Kidney Tubules/metabolism , Magnesium Deficiency/genetics , Nephrocalcinosis/genetics , Animals , Claudins/genetics , Humans , Ion Transport , Magnesium Deficiency/metabolism , Nephrocalcinosis/metabolism , Tight Junctions/metabolism
16.
EBioMedicine ; 50: 366-378, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31735555

ABSTRACT

BACKGROUND: Intrarenal calcium oxalate (CaOx) crystals induce inflammation and kidney tubular cell injury, which are processes that involve TLR4/NF-κB signalling. A recent genome-wide gene expression profile analysis of Randall's plaques in CaOx stone patients revealed that the expression of the long noncoding RNA H19 was significantly upregulated. However, to date, its role in kidney CaOx stones has not been reported. METHOD: A Gene Expression Omnibus (GEO) dataset was utilized to analyse gene expression profiles. Luciferase reporter, Western blotting, qRT-PCR, immunofluorescence staining and reactive oxygen species (ROS) assays were employed to study the molecular mechanism of HMGB1/TLR4/NF-κB regulation by H19 and miR-216b. In vitro and in vivo assays were performed to further confirm the proinflammatory and prooxidative stress effects. FINDING: H19 expression was significantly increased and positively correlated with the expression levels of HMGB1, TLR4 and NF-κB in Randall's plaques and glyoxylate-induced CaOx nephrocalcinosis mouse models. H19 interacted with miR-216b and suppressed its expression. Additionally, miR-216b inhibited HMGB1 expression by directly binding its 3'-untranslated region. Moreover, H19 downregulation inhibited HMGB1, TLR4 and NF-κB expression and suppressed CaOx nephrocalcinosis-induced renal tubular epithelial cell injury, NADPH oxidase, and oxidative stress in vivo and in vitro. Interestingly, miR-216b inhibition partially reversed the inhibitory effect of H19 knockdown on HMGB1 expression. INTERPRETATION: We determined that H19 might serve as a facilitator in the process of CaOx nephrocalcinosis-induced oxidative stress and renal tubular epithelial cell injury, and we revealed that the interaction between H19 and miR-216b could exert its effect via the HMGB1/TLR4/NF-κB pathway. FUNDING: This work was supported by the National Nature Science Foundation of China (Nos. 8196030190, 8190033175, 81370805, 81470935, 81900645, 81500534, and 81602236).


Subject(s)
Calcium Oxalate/metabolism , Epithelial Cells/metabolism , Kidney Tubules/metabolism , Nephrocalcinosis/etiology , Nephrocalcinosis/metabolism , RNA, Long Noncoding/genetics , RNA, Untranslated/genetics , 3' Untranslated Regions , Animals , Biomarkers , Calcium Oxalate/chemistry , Cell Line , Computational Biology , Disease Models, Animal , Epithelial Cells/pathology , Gene Expression Profiling , Gene Expression Regulation , HMGB1 Protein/genetics , Humans , Immunohistochemistry , Kidney Tubules/pathology , Male , Mice , Models, Biological , Nephrocalcinosis/pathology , Oxidative Stress , RNA Interference , Reactive Oxygen Species/metabolism
17.
Am J Physiol Renal Physiol ; 316(5): F966-F969, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30838875

ABSTRACT

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


Subject(s)
Calcium/metabolism , Hypercalciuria/complications , Kidney Calculi/etiology , Kidney Tubules, Proximal/metabolism , Membrane Transport Proteins/metabolism , Nephrocalcinosis/etiology , Renal Reabsorption , Animals , Claudins/metabolism , Humans , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Ion Transport , Kidney Calculi/metabolism , Kidney Calculi/physiopathology , Kidney Tubules, Proximal/physiopathology , Nephrocalcinosis/metabolism , Nephrocalcinosis/physiopathology
18.
J Pediatr ; 203: 391-399.e1, 2018 12.
Article in English | MEDLINE | ID: mdl-30470382

ABSTRACT

OBJECTIVE: To determine whether multiple daily injections of parathyroid hormone (PTH) 1-34 are safe and effective as long-term therapy for children with hypoparathyroidism. STUDY DESIGN: Linear growth, bone accrual, renal function, and mineral homeostasis were studied in a long-term observational study of PTH 1-34 injection therapy in 14 children. METHODS: Subjects were 14 children with hypoparathyroidism attributable to autoimmune polyglandular syndrome type 1 (N = 5, ages 7-12 years) or calcium receptor mutation (N = 9, ages 7-16 years). Mean daily PTH 1-34 dose was 0.75 ± 0.15 µg/kg/day. Treatment duration was 6.9 ± 3.1 years (range 1.5-10 years). Patients were evaluated semiannually at the National Institutes of Health Clinical Center. RESULTS: Mean height velocity and lumbar spine, whole body, and femoral neck bone accretion velocities were normal throughout the study. In the first 2 years, distal one-third radius bone accrual velocity was reduced compared with normal children (P < .003). Serum alkaline phosphatase correlated with PTH 1-34 dose (P < .006) and remained normal (235.3 ± 104.8 [SD] U/L, N: 51-332 U/L). Mean serum and 24-hour urine calcium levels were 2.05 ± 0.11 mmol/L (N: 2.05-2.5 mmol/L) and 6.93 ± 1.3 mmol/24 hour (N: 1.25-7.5 mmol/24 hour), respectively-with fewer high urine calcium levels vs baseline during calcitriol and calcium treatment (P < .001). Nephrocalcinosis progressed in 5 of 12 subjects who had repeated renal imaging although renal function remained normal. CONCLUSIONS: Twice-daily or thrice-daily subcutaneous PTH 1-34 injections provided safe and effective replacement therapy for up to 10 years in children with hypoparathyroidism because of autoimmune polyglandular syndrome type 1 or calcium receptor mutation.


Subject(s)
Body Height/drug effects , Hypoparathyroidism/drug therapy , Parathyroid Hormone/therapeutic use , Adolescent , Calcinosis , Calcium/blood , Calcium/urine , Child , Creatinine/urine , DNA Mutational Analysis , Female , Homeostasis , Hormone Replacement Therapy , Humans , Kidney Function Tests , Linear Models , Male , Nephrocalcinosis/metabolism , Parathyroid Hormone/administration & dosage , Phosphorus/blood , Phosphorus/urine , Polyendocrinopathies, Autoimmune/genetics , Receptors, Calcium-Sensing/genetics , Treatment Outcome , Vitamin D/blood
19.
Sci Rep ; 8(1): 14157, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242165

ABSTRACT

Nephrocalcinosis often begins on a calcium phosphate deposit, at the tip of the medullo-papillary complex (MPC) known as Randall's plaque (RP). Contextualizing proximally observed biominerals within the MPC has led us to postulate a mechanobiological switch that can trigger interstitial biomineralization at the MPC tip, remote from the intratubular biominerals. Micro X-ray computed tomography scans of human MPCs correlated with transmission and scanning electron micrographs, and X-ray energy dispersive spectrometry demonstrated novel findings about anatomically-specific biominerals. An abundance of proximal intratubular biominerals were associated with emergence of distal interstitial RP. The fundamental architecture of the MPC and mineral densities at the proximal and distal locations of the MPC differed markedly. A predominance of plate-like minerals or radially oriented plate-like crystallites within spheroidal minerals in the proximal intratubular locations, and core-shell type crystallites within spheroidal minerals in distal interstitial locations were observed. Based on the MPC anatomic location of structure-specific biominerals, a biological switch within the mineral-free zone occurring between the proximal and distal locations is postulated. The "on" and "off" switch is dependent on changes in the pressure differential resulting from changes in tubule diameters; the "Venturi effect" changes the "circumferential strain" and culminates in interstitial crystal deposits in the distal tubule wall in response to proximal tubular obstruction. These distal interstitial mineralizations can emerge into the collecting system of the kidney linking nephrocalcinosis with nephrolithiasis.


Subject(s)
Biomineralization/physiology , Kidney Medulla/physiology , Calcium Phosphates/metabolism , Humans , Kidney Medulla/metabolism , Minerals/metabolism , Nephrocalcinosis/metabolism , Nephrocalcinosis/physiopathology , X-Ray Microtomography/methods
20.
Curr Opin Nephrol Hypertens ; 27(4): 268-276, 2018 07.
Article in English | MEDLINE | ID: mdl-29782346

ABSTRACT

PURPOSE OF REVIEW: The greatest risk factor for kidney stone formation is increased urinary calcium excretion. Most filtered calcium is reabsorbed from the proximal tubule and the thick ascending limb (TAL) of Henle's loop via a paracellular pathway. Claudins are tight junction proteins that confer the permeability properties of an epithelium. We review the contribution of renal claudins to nephron calcium permeability and how perturbations in these pathways cause alterations in tubular calcium transport, hypercalciuria, nephrocalcinosis, or nephrolithiasis. RECENT FINDINGS: Claudin-16 and Claudin-19 form a complex with claudin-3 enabling divalent cation permeability in the TAL. Claudin-14 interacts with claudin-16 to attenuate calcium permeability through this pore. Intronic mutations in claudin-14 increase expression causing hypercalciuria and kidney stones. A different type of TAL tight junction pore is composed of claudin-10b, which does not preferentially permeate calcium. Deletion of claudin-10b results in increased expression of the claudin-16/claudin-19 complex expressed in the medullary TAL and nephrocalcinosis. SUMMARY: Alterations to claudins expressed in the TAL tight junction greatly affects calcium homeostasis as highlighted by point mutations in claudin-16 or claudin-19 causing FHHNC or gain of function mutations in claudin-14 causing kidney stones.


Subject(s)
Calcium/metabolism , Claudins/metabolism , Kidney Tubules/metabolism , Nephrolithiasis/metabolism , Animals , Cell Membrane Permeability , Humans , Hypercalciuria/metabolism , Ion Transport , Nephrocalcinosis/metabolism , Nephrolithiasis/physiopathology , Tight Junctions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...