Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Urolithiasis ; 50(2): 119-130, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35122487

ABSTRACT

In primary hyperoxaluria, increased hepatic oxalate production sometimes leads to severe nephrocalcinosis and early end-stage kidney disease. Oral administration of Oxalobacter formigenes (O. formigenes), an oxalate-degrading bacterium, is thought to derive oxalate from systemic sources by inducing net enteric oxalate secretion. Here, the impact of O. formigenes on nephrocalcinosis was investigated in an ethylene glycol rat model mimicking hepatic oxalate overproduction in primary hyperoxaluria. Eighteen rats were administered ethylene glycol (0.75% in drinking water) for 6 weeks, of which 9 were treated by oral gavage with O. formigenes and 9 received vehicle. Five control rats did not receive ethylene glycol or O. formigenes. Plasma and urinary oxalate levels, calcium oxalate crystalluria, urinary volume, fluid intake, and serum creatinine were monitored during the study. On killing, nephrocalcinosis was quantified. Ethylene glycol intake induced pronounced hyperoxalemia, hyperoxaluria, calcium oxalate crystalluria and nephrocalcinosis. Concomitant O. formigenes treatment partially prevented the ethylene glycol-induced increase in plasma oxalate and completely prevented nephrocalcinosis. Urinary oxalate excretion was not reduced by O. formigenes treatment. Nevertheless, absence of crystals in renal tissue of O. formigenes-treated ethylene glycol animals indicates that the propensity for oxalate to crystallize in the kidneys was reduced compared to non-treated animals. This is supported by the lower plasma oxalate concentrations in O. formigenes-treated animals. This study shows a beneficial effect of O. formigenes treatment on ethylene glycol-induced hyperoxalemia and nephrocalcinosis, and thus supports a possible beneficial effect of O. formigenes in primary hyperoxaluria.


Subject(s)
Hyperoxaluria, Primary , Hyperoxaluria , Nephrocalcinosis , Animals , Calcium Oxalate , Humans , Hyperoxaluria/complications , Hyperoxaluria, Primary/complications , Nephrocalcinosis/complications , Nephrocalcinosis/prevention & control , Oxalates/urine , Oxalobacter formigenes , Rats
2.
Surg Today ; 52(1): 12-21, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33464414

ABSTRACT

Short-bowel syndrome (SBS) is defined as a state of malabsorption after resection or loss of a major portion of the bowel due to congenital or acquired factors. This article presents an overview on the recent management of pediatric SBS. The pediatric SBS population is very heterogeneous. The incidence of SBS is estimated to be 24.5 per 100,000 live births. The nutritional, medical, and surgical therapies available require a comprehensive evaluation. Thus, multidisciplinary intestinal rehabilitation programs (IRPs) are necessary for the management of these complex patients. The key points of focus in IRP management are hepato-protective strategies to minimize intestinal failure-associated liver disease; the aggressive prevention of catheter-related bloodstream infections; strategic nutritional supply to optimize the absorption of enteral calories; and the management and prevention of small bowel bacterial overgrowth, nephrocalcinosis, and metabolic bone disease. As the survival rate of children with SBS currently exceeds 90%, the application of small bowel transplantation has been evolving. The introduction of innovative treatments, such as combined therapy of intestinotrophic hormones, including glucagon-like peptide-2, may lead to further improvements in patients' quality of life.


Subject(s)
Short Bowel Syndrome/rehabilitation , Short Bowel Syndrome/therapy , Age Factors , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/prevention & control , Child, Preschool , Female , Glucagon-Like Peptide 2/therapeutic use , Humans , Incidence , Infant , Infant, Newborn , Intestinal Failure/etiology , Intestinal Failure/prevention & control , Intestine, Small/transplantation , Liver Diseases/etiology , Liver Diseases/prevention & control , Male , Nephrocalcinosis/etiology , Nephrocalcinosis/prevention & control , Quality of Life , Short Bowel Syndrome/epidemiology , Short Bowel Syndrome/etiology
3.
Endocrinol Diabetes Metab ; 4(2): e00185, 2021 04.
Article in English | MEDLINE | ID: mdl-33855198

ABSTRACT

There is a paucity of data on the use of SGLT2 inhibitors on outcomes in kidney transplant recipients. There may be concern in initiating these agents, especially within the first year post-transplant when renal function is more labile and immunosuppression more intense, due to a presumed high risk of urinary infections and acute kidney injury. This is a retrospective study on 50 kidney transplant recipients, half of whom were started on therapy within the first year of transplant. Over a follow-up period of 6 months, overall patients had a statistically significant improvement in weight by -2.95 kg [SD 3.54, P = <.0001 (CI: 3.53, 1.50)] as well as hypomagnesemia 0.13 [SD 1.73, P = .0004 (CI: 0.06, 0.20)]. Overall insulin usage declined by -3.7 units [SD 22.8, P = .17]. 14% of patients had at least one urinary tract infection although this rate is not different (~20%) than that reported historically in this high-risk population.


Subject(s)
Diabetic Nephropathies/metabolism , Diabetic Nephropathies/surgery , Electrolytes/metabolism , Kidney Transplantation , Sodium-Glucose Transporter 2 Inhibitors/administration & dosage , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Adult , Aged , Female , Follow-Up Studies , Humans , Hypercalciuria/etiology , Hypercalciuria/prevention & control , Kidney Transplantation/adverse effects , Male , Middle Aged , Nephrocalcinosis/etiology , Nephrocalcinosis/prevention & control , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Renal Tubular Transport, Inborn Errors/etiology , Renal Tubular Transport, Inborn Errors/prevention & control , Retrospective Studies , Risk , Time Factors , Treatment Outcome , Weight Gain
4.
J Pediatr Endocrinol Metab ; 34(6): 791-798, 2021 Jun 25.
Article in English | MEDLINE | ID: mdl-33837680

ABSTRACT

OBJECTIVES: X-linked hypophosphatemic rickets (XLH) is a congenital fibroblast growth factor (FGF)23-related metabolic bone disease that is treated with active vitamin D and phosphate as conventional therapies. Complications of these therapies include nephrocalcinosis (NC) caused by excessive urine calcium and phosphate concentrations. Recently, an anti-FGF23 antibody, burosumab, was developed and reported to be effective in poorly-controlled or severe XLH patients. This study aimed to reveal the impact of switching treatments in relatively well-controlled XLH children with the Rickets Severity Scale less than 2.0. METHODS: The effects of the two treatments in eight relatively well-controlled XLH children with a mean age of 10.4 ± 1.9 years were compared retrospectively for the same treatment duration (31 ± 11 months) before and after the baseline. RESULTS: Actual doses of alfacalcidol and phosphate as conventional therapy were 150.9 ± 43.9 ng/kg and 27.5 ± 6.3 mg/kg per day, respectively. Renal echography revealed spotty NC in 8/8 patients, but no aggravation of NC was detected by switching treatments. Switching treatments increased TmP/GFR (p=0.002) and %TRP (p<0.001), and improved the high urine calcium/creatinine ratio to the normal range (p<0.001) although both treatments controlled disease markers equally. Additionally, low intact parathyroid hormone during conventional therapy was increased within the normal range by switching treatments. CONCLUSIONS: Our results suggest that a high dose of alfacalcidol was needed to control the disease, but it caused hypercalciuria and NC. We concluded that switching treatments in relatively well-controlled XLH children improved renal phosphate reabsorption and decreased urine calcium extraction, and may have the potential to prevent NC.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Drug Substitution/methods , Familial Hypophosphatemic Rickets/drug therapy , Fibroblast Growth Factors/antagonists & inhibitors , Hydroxycholecalciferols/therapeutic use , Nephrocalcinosis/prevention & control , Bone Density Conservation Agents/therapeutic use , Child , Familial Hypophosphatemic Rickets/pathology , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/immunology , Follow-Up Studies , Humans , Injections, Intravenous , Male , Prognosis , Retrospective Studies
5.
Cancer Chemother Pharmacol ; 86(3): 383-391, 2020 09.
Article in English | MEDLINE | ID: mdl-32789758

ABSTRACT

PURPOSE: Cetuximab and panitumumab are monoclonal antibodies that target the epidermal growth factor receptor (EGFR). Treatment with cetuximab and panitumumab commonly causes hypomagnesemia, and optimal management of this adverse effect remains unclear. Here, we evaluated the optimal magnesium replacement points based on the risk of severe hypomagnesemia in colorectal cancer patients who received cetuximab or panitumumab. METHODS: We retrospectively evaluated 184 patients who received cetuximab or panitumumab for colorectal cancer at Ogaki Municipal Hospital (Ogaki, Japan) between January 2010 and December 2019. Univariate analyses were conducted to evaluate the relationship between patient baseline characteristics and development of hypomagnesemia following cetuximab or panitumumab treatment. Variables that were significantly associated with hypomagnesemia in the univariate analyses as well as previously reported risk factors were entered into a multivariate logistic regression model. RESULTS: The incidence of hypomagnesemia was associated with panitumumab treatment, pre-replenishment serum magnesium concentration, treatment duration, and treatment line. Severe hypomagnesemia post-cetuximab or panitumumab treatment was significantly associated with low baseline magnesium concentrations (< 1.8 mg/dL; odds ratio 18.100, 95% confidence interval 1.570-210.000; p = 0.020) and low serum magnesium concentrations during treatment (< 1.1 mg/dL; odds ratio 93.800, 95% confidence interval 3.510-2510.000; p = 0.007). CONCLUSION: To minimize the risk of severe hypomagnesemia during anti-EGFR treatment, magnesium replenishment should be initiated in patients with pre-replenishment concentrations of < 1.8 mg/dL, preferably before reaching intra-treatment concentrations of < 1.1 mg/dL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Colorectal Neoplasms/drug therapy , Hypercalciuria/prevention & control , Magnesium/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Nephrocalcinosis/prevention & control , Renal Tubular Transport, Inborn Errors/prevention & control , Adult , Aged , Aged, 80 and over , Cetuximab/administration & dosage , Colorectal Neoplasms/pathology , Female , Follow-Up Studies , Humans , Hypercalciuria/chemically induced , Hypercalciuria/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Nephrocalcinosis/chemically induced , Nephrocalcinosis/pathology , Panitumumab/administration & dosage , Prognosis , Renal Tubular Transport, Inborn Errors/chemically induced , Renal Tubular Transport, Inborn Errors/pathology , Retrospective Studies
7.
Exp Cell Res ; 376(2): 210-220, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30690028

ABSTRACT

Cetuximab, an inhibitor of the epidermal growth factor receptor that is used widely to treat human cancers including oral squamous cell carcinoma (OSCC), has characteristic side effects of skin rash and hypomagnesemia. However, the mechanisms of and therapeutic agents for skin rashes and hypomagnesemia are still poorly understood. Our gene expression profiling analyses showed that cetuximab activates the p38 MAPK pathways in human skin cells (human keratinocyte cell line [HaCaT]) and inhibits c-Fos-related signals in human embryonic kidney cells (HEK293). We found that while the p38 inhibitor SB203580 inhibited the expression of p38 MAPK targets in HaCaT cells, flavagline reactivated c-Fos-related factors in HEK293 cells. It is noteworthy that, in addition to not interfering with the effect of cetuximab by both compounds, flavagline has additive effect for OSCC growth inhibition in vivo. Collectively, our results indicate that combination of cetuximab and these potential therapeutic agents for cetuximab-related toxicities could be a promising therapeutic strategy for patients with OSCC.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Carcinoma, Squamous Cell/drug therapy , Cetuximab/adverse effects , Growth Inhibitors/therapeutic use , Imidazoles/therapeutic use , Mouth Neoplasms/drug therapy , Pyridines/therapeutic use , Animals , Carcinoma, Squamous Cell/complications , Cell Line, Tumor , Drug Therapy, Combination , ErbB Receptors/antagonists & inhibitors , Exanthema/chemically induced , Exanthema/genetics , Exanthema/prevention & control , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Growth Inhibitors/adverse effects , Growth Inhibitors/antagonists & inhibitors , HEK293 Cells , Humans , Hypercalciuria/chemically induced , Hypercalciuria/genetics , Hypercalciuria/prevention & control , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/complications , Mouth Neoplasms/genetics , Nephrocalcinosis/chemically induced , Nephrocalcinosis/genetics , Nephrocalcinosis/prevention & control , Renal Tubular Transport, Inborn Errors/chemically induced , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/prevention & control , Transcriptome , Xenograft Model Antitumor Assays
8.
Eur J Med Genet ; 62(11): 103577, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30423445

ABSTRACT

Loss-of-function mutations in CYP24A1 (MIM 126065 20q13.2), the gene encoding the 24-hydroxylase responsible for 25-OH-D and 1,25-(OH)2D degradation, are identified in about 20% of patients presenting Idiopathic Infantile Hypercalcemia (IIH) (MIM 143880). Common features of this autosomal recessive condition included hypercalcemia with hypercalciuria, suppressed PTH and a high 25-OH-D3:24,25-(OH)2D3 ratio. Medical care mainly relies on sun protection and life-long contraindication of vitamin D to avoid complications such as early nephrocalcinosis and renal failure. Molecular diagnosis therefore keeps a crucial place in the diagnosis of IIH, and genetic counseling should be systematically recommended to prevent vitamin D administration in affected siblings. In this report is described the molecular characterization of a CYP24A1 deletion identified in two unrelated families. This highlights the potential role of CYP24A1 copy number variations (CNV) in IIH. Considering the presence of CNV affecting CYP24A1 in public databases, CNV analysis should be systematically added to the sequencing studies in IIH. Targeted Massively Parallel Sequencing (MPS) study coupled with a CNV detection tool called CovCop is a powerful method to detect genic rearrangement and improve genetic analysis.


Subject(s)
Hypercalcemia/diagnosis , Hypercalcemia/genetics , Infant, Newborn, Diseases/diagnosis , Infant, Newborn, Diseases/genetics , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/genetics , Pathology, Molecular , Vitamin D3 24-Hydroxylase/genetics , Adolescent , Child , Child, Preschool , DNA Copy Number Variations/genetics , Female , Humans , Hypercalcemia/drug therapy , Hypercalcemia/pathology , Infant , Infant, Newborn, Diseases/drug therapy , Infant, Newborn, Diseases/pathology , Loss of Function Mutation/genetics , Male , Metabolism, Inborn Errors/drug therapy , Metabolism, Inborn Errors/pathology , Nephrocalcinosis/drug therapy , Nephrocalcinosis/genetics , Nephrocalcinosis/prevention & control , Renal Insufficiency/drug therapy , Renal Insufficiency/genetics , Renal Insufficiency/prevention & control , Sequence Deletion/genetics , Vitamin D/therapeutic use
9.
Urolithiasis ; 47(1): 79-89, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30535828

ABSTRACT

Primary hyperoxaluria (PH) is caused by genetic mutations resulting in oxalate overproduction leading to nephrolithiasis, nephrocalcinosis, extrarenal manifestations, chronic kidney disease, and end-stage renal disease. Advances in genetic testing techniques have improved our ability to efficiently and effectively obtain a definitive diagnosis of PH as well as easily screen at-risk family members. Similarly, advances in technologies related to intervening at the genetic and molecular level promise to change the way we treat patients with PH. In this review, we provide an update regarding the identification of underlying molecular and biochemical causes of inherited hyperoxalurias, clinical manifestations, and treatment strategies.


Subject(s)
Genetic Testing/methods , Hyperoxaluria, Primary/genetics , Kidney Calculi/prevention & control , Nephrocalcinosis/prevention & control , Oxalates/metabolism , Alcohol Oxidoreductases/genetics , Fluid Therapy , Humans , Hyperoxaluria, Primary/complications , Hyperoxaluria, Primary/diagnosis , Hyperoxaluria, Primary/therapy , Kidney Calculi/etiology , Kidney Calculi/surgery , Kidney Transplantation , Lithotripsy , Liver Transplantation , Mutation , Nephrocalcinosis/etiology , Oxo-Acid-Lyases/genetics , Pyridoxine/therapeutic use , Renal Replacement Therapy/methods , Transaminases/genetics , Treatment Outcome
10.
Nat Commun ; 9(1): 5454, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30575740

ABSTRACT

CRISPR/Cas9 technology offers novel approaches for the development of new therapies for many unmet clinical needs, including a significant number of inherited monogenic diseases. However, in vivo correction of disease-causing genes is still inefficient, especially for those diseases without selective advantage for corrected cells. We reasoned that substrate reduction therapies (SRT) targeting non-essential enzymes could provide an attractive alternative. Here we evaluate the therapeutic efficacy of an in vivo CRISPR/Cas9-mediated SRT to treat primary hyperoxaluria type I (PH1), a rare inborn dysfunction in glyoxylate metabolism that results in excessive hepatic oxalate production causing end-stage renal disease. A single systemic administration of an AAV8-CRISPR/Cas9 vector targeting glycolate oxidase, prevents oxalate overproduction and kidney damage, with no signs of toxicity in Agxt1-/- mice. Our results reveal that CRISPR/Cas9-mediated SRT represents a promising therapeutic option for PH1 that can be potentially applied to other metabolic diseases caused by the accumulation of toxic metabolites.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , CRISPR-Cas Systems , Genetic Therapy/methods , Hyperoxaluria, Primary/therapy , Oxalates/urine , Alcohol Oxidoreductases/genetics , Animals , Disease Models, Animal , Gene Editing , HEK293 Cells , Humans , Male , Mice , Nephrocalcinosis/prevention & control
11.
J Trace Elem Med Biol ; 50: 327-331, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30262299

ABSTRACT

In advanced squamous cell carcinoma of the head and neck, concomitant radiotherapy with cisplatin and/or cetuximab is frequently combined with cisplatin-based induction chemotherapy, which can cause severe hypomagnesemia, hypocalcemia, and hypokalemia. The aim of our study was to analyze the effects of magnesium sulfate supplementation on the incidence of hypomagnesemia, hypokalemia, and hypocalcemia during four cycles of TPF (docetaxel, cisplatin, and 5-fluorouracil) induction chemotherapy followed by concomitant radiotherapy (CRT) with cisplatin and cetuximab. Twenty-five patients included in a phase II prospective study received routine magnesium sulfate infusions before each cycle of cisplatin, and additional supplementation based on laboratory findings. During TPF, the incidence of grade 1/2 and grade 3/4 hypomagnesemia was 16% and 4%, respectively; and increased despite magnesium supplementation during CRT to 72% and 8%, respectively. During TPF, a grade 2 and grade 4 hypocalcemia occurred in 8% and 4%, respectively; and during CRT, it reached 36% (grade 1/2). Grade 1 hypokalemia only was observed during TPF (4%) and CRT (8%). The median amounts of supplemented magnesium sulfate during TPF and CRT were 20 mEq and 50 mEq, respectively. It appears that a low incidence of grade 3/4 hypomagnesemia and hypocalcemia in our patients resulted from intensive magnesium supplementation. Thorough measurements of magnesium and calcium during cisplatin-based chemoradiation protocols in patients with head and neck cancer are crucial in preventing the development of grade 3/4 hypomagnesemia and hypocalcemia.


Subject(s)
Chemoradiotherapy/adverse effects , Hypercalciuria/prevention & control , Hypocalcemia/prevention & control , Magnesium Sulfate/therapeutic use , Nephrocalcinosis/prevention & control , Renal Tubular Transport, Inborn Errors/prevention & control , Cetuximab/adverse effects , Cetuximab/therapeutic use , Cisplatin/adverse effects , Cisplatin/therapeutic use , Female , Head and Neck Neoplasms/drug therapy , Humans , Male , Prospective Studies
12.
J Am Soc Nephrol ; 29(6): 1636-1648, 2018 06.
Article in English | MEDLINE | ID: mdl-29654213

ABSTRACT

Background The high cardiovascular morbidity and mortality of patients with CKD may result in large part from medial vascular calcification, a process promoted by hyperphosphatemia and involving osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs). Reduced serum zinc levels have frequently been observed in patients with CKD, but the functional relevance of this remains unclear.Methods We performed experiments in primary human aortic VSMCs; klotho-hypomorphic (kl/kl), subtotal nephrectomy, and cholecalciferol-overload mouse calcification models; and serum samples from patients with CKD.Results In cultured VSMCs, treatment with zinc sulfate (ZnSO4) blunted phosphate-induced calcification, osteo-/chondrogenic signaling, and NF-κB activation. ZnSO4 increased the abundance of zinc-finger protein TNF-α-induced protein 3 (TNFAIP3, also known as A20), a suppressor of the NF-κB pathway, by zinc-sensing receptor ZnR/GPR39-dependent upregulation of TNFAIP3 gene expression. Silencing of TNFAIP3 in VSMCs blunted the anticalcific effects of ZnSO4 under high phosphate conditions. kl/kl mice showed reduced plasma zinc levels, and ZnSO4 supplementation strongly blunted vascular calcification and aortic osteoinduction and upregulated aortic Tnfaip3 expression. ZnSO4 ameliorated vascular calcification in mice with chronic renal failure and mice with cholecalciferol overload. In patients with CKD, serum zinc concentrations inversely correlated with serum calcification propensity. Finally, ZnSO4 ameliorated the osteoinductive effects of uremic serum in VSMCs.Conclusions Zinc supplementation ameliorates phosphate-induced osteo-/chondrogenic transdifferentiation of VSMCs and vascular calcification through an active cellular mechanism resulting from GPR39-dependent induction of TNFAIP3 and subsequent suppression of the NF-κB pathway. Zinc supplementation may be a simple treatment to reduce the burden of vascular calcification in CKD.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Kidney Failure, Chronic/blood , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Vascular Calcification/prevention & control , Zinc Sulfate/pharmacology , Animals , Aorta , Cell Transdifferentiation , Cells, Cultured , Dietary Supplements , Disease Models, Animal , Gene Expression/drug effects , Gene Silencing , Glucuronidase/genetics , Humans , Hydroxyethylrutoside , Hyperphosphatemia/blood , Hyperphosphatemia/complications , Klotho Proteins , Mice , NF-kappa B/antagonists & inhibitors , Nephrectomy , Nephrocalcinosis/prevention & control , Phosphates , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Vascular Calcification/blood , Vascular Calcification/etiology , Zinc/blood
13.
Kidney Int ; 93(3): 580-588, 2018 03.
Article in English | MEDLINE | ID: mdl-29129401

ABSTRACT

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


Subject(s)
Claudins/deficiency , Hypercalciuria/prevention & control , Kidney Tubules, Distal/metabolism , Loop of Henle/metabolism , Magnesium Deficiency/prevention & control , Animals , Calcium/metabolism , Claudins/genetics , Disease Models, Animal , Gene Deletion , Genetic Predisposition to Disease , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Kidney Tubules, Distal/pathology , Kidney Tubules, Distal/physiopathology , Loop of Henle/pathology , Loop of Henle/physiopathology , Magnesium/metabolism , Magnesium Deficiency/genetics , Magnesium Deficiency/metabolism , Magnesium Deficiency/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Nephrocalcinosis/genetics , Nephrocalcinosis/metabolism , Nephrocalcinosis/physiopathology , Nephrocalcinosis/prevention & control , Phenotype , Sodium/metabolism
14.
Kidney Int ; 93(3): 656-669, 2018 03.
Article in English | MEDLINE | ID: mdl-29241624

ABSTRACT

Primary/secondary hyperoxalurias involve nephrocalcinosis-related chronic kidney disease (CKD) leading to end-stage kidney disease. Mechanistically, intrarenal calcium oxalate crystal deposition is thought to elicit inflammation, tubular injury and atrophy, involving the NLRP3 inflammasome. Here, we found that mice deficient in NLRP3 and ASC adaptor protein failed to develop nephrocalcinosis, compromising conclusions on nephrocalcinosis-related CKD. In contrast, hyperoxaluric wild-type mice developed profound nephrocalcinosis. NLRP3 inhibition using the ß-hydroxybutyrate precursor 1,3-butanediol protected such mice from nephrocalcinosis-related CKD. Interestingly, the IL-1 inhibitor anakinra had no such effect, suggesting IL-1-independent functions of NLRP3. NLRP3 inhibition using 1,3-butanediol treatment induced a shift of infiltrating renal macrophages from pro-inflammatory (CD45+F4/80+CD11b+CX3CR1+CD206-) and pro-fibrotic (CD45+F4/80+CD11b+CX3CR1+CD206+TGFß+) to an anti-inflammatory (CD45+F4/80+CD11b+CD206+TGFß-) phenotype, and prevented renal fibrosis. Finally, in vitro studies with primary murine fibroblasts confirmed the non-redundant role of NLRP3 in the TGF-ß signaling pathway for fibroblast activation and proliferation independent of the NLRP3 inflammasome complex formation. Thus, nephrocalcinosis-related CKD involves NLRP3 but not necessarily via intrarenal IL-1 release but rather via other biological functions including TGFR signaling and macrophage polarization. Hence, NLRP3 may be a promising therapeutic target in hyperoxaluria and nephrocalcinosis.


Subject(s)
Cell Plasticity , Hyperoxaluria/metabolism , Inflammasomes/metabolism , Interleukin-1/metabolism , Kidney/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephrocalcinosis/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Butylene Glycols/pharmacology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Cell Plasticity/drug effects , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Hyperoxaluria/drug therapy , Hyperoxaluria/immunology , Hyperoxaluria/pathology , Inflammasomes/drug effects , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-1/immunology , Kidney/immunology , Kidney/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nephrocalcinosis/immunology , Nephrocalcinosis/pathology , Nephrocalcinosis/prevention & control , Phenotype , Receptors, Transforming Growth Factor beta/metabolism , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/prevention & control , Signal Transduction
15.
Trials ; 18(1): 424, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28893306

ABSTRACT

BACKGROUND: The pivotal role of vitamin D (vit D) in skeletal health is well known. Neonatal vit D storage at birth is dependent on maternal levels, and newborns receive 50-70% of their mother's 25-hydroxyvitamin D [25(OH)D]. Deficiency of vit D can lead to prematurity bone disease, with an incidence of up to 55% in infants weighing < 1000 g. The aim of this study is to assess the effectiveness of monitored supplementation of vit D in a population of preterm infants. METHODS/DESIGN: Preterm infants born at 24-32 weeks of gestation will be recruited within the first 7 days of life. Depending on the type of feeding, and after reaching partial enteral feeding or at 7 days of life, vit D supplementation will consist of 500 IU and an additional 150-300 IU/kg included in human milk fortifiers (if fed exclusively with breast milk) or 190 IU/kg in milk formulas. Subjects will be randomised to either monitored (with an option of dose modification based on 25(OH)D levels as per protocol) or standard therapy up to 52 weeks of post-conceptional age (PCA). The primary outcome measure will be the number of neonates with deficiency or excess levels of 25(OH)D at 40 ±2 weeks of PCA. Additional 25(OH)D levels will be measured at birth, at 4 and 8 weeks of age, and/or at 35 and 52 ±2 weeks of PCA. Secondary objectives will include the incidence of osteopenia, nephrocalcinosis and nephrolithiasis. Serum parameters of calcium phosphorus metabolism will also be measured. DISCUSSION: Despite multiple years of research and numerous publications, there is still a lack of consensus in regard to how much vit D infants should receive and how long they should receive it. Because 80% of calcium and phosphorus placental transfer occurs between 24 and 40 weeks of gestation, preterm infants are especially prone to adverse effects of vit D insufficiency. However, both inadequate and excessive amounts of vit D may be unsafe and lead to serious health issues. The results of our study may shed new light on these concerns and contribute to optimising vit D supplementation. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03087149 . Registered on 15 March 2017.


Subject(s)
Cholecalciferol/administration & dosage , Dietary Supplements , Infant, Premature , Premature Birth , Vitamin D Deficiency/drug therapy , Biomarkers/blood , Bone Diseases, Metabolic/epidemiology , Bone Diseases, Metabolic/prevention & control , Cholecalciferol/adverse effects , Clinical Protocols , Dietary Supplements/adverse effects , Gestational Age , Humans , Incidence , Infant , Infant, Newborn , Infant, Premature/blood , Nephrocalcinosis/epidemiology , Nephrocalcinosis/prevention & control , Nephrolithiasis/epidemiology , Nephrolithiasis/prevention & control , Poland/epidemiology , Premature Birth/blood , Research Design , Time Factors , Treatment Outcome , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamin D Deficiency/blood , Vitamin D Deficiency/diagnosis , Vitamin D Deficiency/epidemiology
16.
Pediatr Diabetes ; 18(5): 327-331, 2017 08.
Article in English | MEDLINE | ID: mdl-28568353

ABSTRACT

Prior to 1971, type Ia glycogen storage disease was marked by life-threatening hypoglycemia, lactic acidosis, severe failure to thrive, and developmental delay. With the introduction of continuous feeds in the 1970s and cornstarch in the 1980s, the prognosis improved, but complications almost universally developed. Changes in the management of type Ia glycogen storage disease have resulted in improved metabolic control, and this manuscript reviews the increasing evidence that complications can be delayed or prevented with optimal metabolic control as previously was seen in diabetes.


Subject(s)
Glycogen Storage Disease Type I/therapy , Precision Medicine , Renal Insufficiency/prevention & control , Adenoma/complications , Adenoma/prevention & control , Adult , Child , Combined Modality Therapy , Glycogen Storage Disease Type I/blood , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/diagnosis , Humans , Hypoglycemia/complications , Hypoglycemia/prevention & control , Liver Neoplasms/complications , Liver Neoplasms/prevention & control , Nephrocalcinosis/complications , Nephrocalcinosis/prevention & control , Nephrolithiasis/complications , Nephrolithiasis/prevention & control , Osteoporosis/complications , Osteoporosis/prevention & control , Prognosis , Renal Insufficiency/complications
17.
Yakugaku Zasshi ; 137(1): 79-82, 2017.
Article in Japanese | MEDLINE | ID: mdl-28049899

ABSTRACT

Hypomagnesemia is one side effect in patients receiving cisplatin. However, there are few reports of cisplatin-induced hypomagnesemia in Japan. We retrospectively investigated the incidence of hypomagnesemia and nephrotoxicity in patients undergoing radiation therapy who were treated with cisplatin alone (dosage: 40 mg/m2, administration interval: 1 week) for cervical cancer. Thirty-two patients undergoing radiation therapy who received cisplatin alone for cervical cancer between January 2012 and May 2016 at Aichi Medical University Hospital were included. We measured patients' serum magnesium and creatinine levels on the day before cisplatin was administered. We utilized the RIFLE criteria (categorized into "risk", "injury", "failure", "loss", and "end-stage kidney disease") to define levels of cisplatin-induced nephrotoxicity, and classified cisplatin-induced nephrotoxicity into "risk" or "injury". Eighteen patients (56.3%) had cisplatin-induced hypomagnesemia, the majority of which occurred after the 4th treatment cycle. The number of patients with moderate renal dysfunction classified as "risk" in the hypomagnesemia group was not significantly higher than in the non-hypomagnesemia group (hypomagnesemia group=27.8%, non-hypomagnesemia group=7.1%; p=0.20). This survey sheds light on the incidence rates of cisplatin-induced hypomagnesemia in patients receiving cisplatin alone. We recommend monitoring the serum magnesium levels during cisplatin administration to prevent hypomagnesemia.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Hypercalciuria/chemically induced , Hypercalciuria/epidemiology , Nephrocalcinosis/chemically induced , Nephrocalcinosis/epidemiology , Renal Tubular Transport, Inborn Errors/chemically induced , Renal Tubular Transport, Inborn Errors/epidemiology , Uterine Cervical Neoplasms/drug therapy , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Combined Modality Therapy , Female , Humans , Hypercalciuria/prevention & control , Incidence , Monitoring, Physiologic , Nephrocalcinosis/prevention & control , Renal Tubular Transport, Inborn Errors/prevention & control , Retrospective Studies , Uterine Cervical Neoplasms/radiotherapy
18.
Endocr J ; 64(3): 283-289, 2017 Mar 31.
Article in English | MEDLINE | ID: mdl-28025445

ABSTRACT

X-linked hypophosphatemia (XLH) is a group of rare disorders caused by defective proximal tubular reabsorption of phosphate. Mutations in the PHEX gene are responsible for the majority of cases. There are very few reports of long-term complications of XLH other than skeletal and dental diseases. The aim of this study was to identify the phenotypic presentation of XLH during adulthood including complications other than skeletal and dental diseases. The clinical and biochemical phenotype of 22 adult patients with a PHEX gene mutation were examined retrospectively from their medical records. 6 patients had hypertension. The average age of hypertension onset was 29.0 years. Secondary hyperparathyroidism preceded the development of hypertension in 5 patients. 1 patient developed tertiary hyperparathyroidism. 15 patients had nephrocalcinosis. 2 patients had chronic renal dysfunction. Patients with hypertension had a significantly lower eGFR (p=0.010) compared to patients without hypertension. No significant difference was found in any other parameters. To examine the genotype-phenotype correlation, 10 adult males were chosen for analysis. No significant genotype-phenotype correlation analysis was revealed in any of the complications. However, there was a possibility that the age at nephrocalcinosis onset was younger in the non-missense mutation group than in the missense mutation group (p=0.063). This study corroborated the view that early-onset hypertension could be one of the characteristic complications seen in XLH patients. Considering the limited number of our patients, further study is necessary to address a potential cause of hypertension. XLH patients require careful lifelong treatment.


Subject(s)
Familial Hypophosphatemic Rickets/physiopathology , Hyperparathyroidism, Secondary/etiology , Hypertension/etiology , Nephrocalcinosis/etiology , Adolescent , Adult , Age of Onset , Bone Density Conservation Agents/therapeutic use , Child , Child, Preschool , Dietary Supplements , Familial Hypophosphatemic Rickets/diet therapy , Familial Hypophosphatemic Rickets/genetics , Female , Hospitals, Pediatric , Humans , Hydroxycholecalciferols/therapeutic use , Hyperparathyroidism, Secondary/epidemiology , Hyperparathyroidism, Secondary/prevention & control , Hypertension/epidemiology , Hypertension/prevention & control , Male , Medical Records , Mutation , Nephrocalcinosis/epidemiology , Nephrocalcinosis/prevention & control , PHEX Phosphate Regulating Neutral Endopeptidase/genetics , Phosphates/therapeutic use , Prevalence , Retrospective Studies , Tokyo/epidemiology , Young Adult
20.
Kidney Int ; 89(5): 1027-1036, 2016 May.
Article in English | MEDLINE | ID: mdl-27083280

ABSTRACT

Pathologic calcification is a significant cause of increased morbidity and mortality in patients with chronic kidney disease. The precise mechanisms of ectopic calcification are not fully elucidated, but it is known to be caused by an imbalance of procalcific and anticalcific factors. In the chronic kidney disease population, an elevated phosphate burden is both highly prevalent and a known risk factor for ectopic calcification. Here we tested whether osteopontin, an inhibitor of calcification, protects against high phosphate load-induced nephrocalcinosis and vascular calcification. Osteopontin knockout mice were placed on a high phosphate diet for 11 weeks. Osteopontin deficiency together with phosphate overload caused uremia, nephrocalcinosis characterized by substantial renal tubular and interstitial calcium deposition, and marked vascular calcification when compared with control mice. Although the osteopontin-deficient mice did not exhibit hypercalcemia or hyperphosphatemia, they did show abnormalities in the mineral metabolism hormone fibroblast growth factor-23. Thus, endogenous osteopontin plays a critical role in the prevention of phosphate-induced nephrocalcinosis and vascular calcification in response to high phosphate load. A better understanding of osteopontin's role in phosphate-induced calcification will hopefully lead to better biomarkers and therapies for this disease, especially in patients with chronic kidney disease and other at-risk populations.


Subject(s)
Aorta, Abdominal/metabolism , Aorta, Thoracic/metabolism , Aortic Diseases/prevention & control , Kidney/metabolism , Nephrocalcinosis/prevention & control , Osteopontin/metabolism , Phosphates , Vascular Calcification/prevention & control , Animals , Aorta, Abdominal/pathology , Aorta, Thoracic/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Biomarkers/blood , Bone Remodeling , Diet , Disease Models, Animal , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Kidney/pathology , Mice, Inbred DBA , Mice, Knockout , Nephrocalcinosis/genetics , Nephrocalcinosis/metabolism , Nephrocalcinosis/pathology , Osteopontin/deficiency , Osteopontin/genetics , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...