Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
J Infect Chemother ; 28(2): 299-303, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34756827

ABSTRACT

Pulmonary oxalosis can be fatal, and Aspergillus tubingensis is commonly resistant to azoles in Japan. We report a case of bronchopulmonary oxalosis caused by A. tubingensis in a non-neutropenic patient who was successfully treated with voriconazole monotherapy. The susceptibility of the isolates to voriconazole and the effective elimination of contagious necrotic tissue by expectoration seemed to be two major factors contributing to the patient's survival. According to the literature review, pulmonary oxalosis is associated with a high mortality rate over a short term. An exploration of detailed information about the genomic characteristics and drug susceptibility of Aspergillus isolates is important for the development of treatment strategies for this life-threatening disease.


Subject(s)
Antifungal Agents , Hyperoxaluria , Antifungal Agents/therapeutic use , Aspergillus/genetics , Humans , Hyperoxaluria/drug therapy , Microbial Sensitivity Tests
2.
Medicina (Kaunas) ; 57(6)2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34204866

ABSTRACT

Background and Objectives: The most common kidney stones are calcium stones and calcium oxalate (CaOx) stones are the most common type of calcium stones. Hyperoxaluria is an essential risk factor for the formation of these stones. Quercetin is a polyphenol with antioxidant, anti-inflammatory, and many other physiological effects. The aim of this study was to investigate the protective effect of quercetin in hyperoxaluria-induced nephrolithiasis. Materials and Methods: Male Wistar-Albino rats weighing 250-300 g (n = 24) were randomized into three groups: Control (n = 8), ethylene glycol (EG) (n = 8), and EG + quercetin (n = 8). One percent EG-water solution was given to all rats except for the control group as drinking water for five weeks. Quercetin-water solution was given to the EG + quercetin group by oral gavage at a dose of 10 mg/kg/day. Malondialdehyde (MDA), catalase (CAT), urea, calcium, and oxalate levels were analyzed in blood and urine samples. Histopathological assessments and immunohistochemical analyses for oxidative stress and inflammation indicators p38 mitogen-activated protein kinase (p38-MAPK) and nuclear factor kappa B (NF-kB) were performed on renal tissues. Results: The MDA levels were significantly lower in the quercetin-treated group than in the EG-treated group (p = 0.001). Although CAT levels were higher in the quercetin-treated group than the EG-administered group, they were not significantly different between these groups. The expression of p38 MAPK was significantly less in the quercetin-treated group than the EG group (p < 0.004). There was no statistically significant difference between the quercetin and EG groups in terms of NF-kB expression. Conclusions: We conclude that hyperoxaluria activated the signaling pathways, which facilitate the oxidative processes leading to oxalate stone formation in the kidneys. Our findings indicated that quercetin reduced damage due to hyperoxaluria. These results imply that quercetin can be considered a therapeutic agent for decreasing oxalate stone formation, especially in patients with recurrent stones due to hyperoxaluria.


Subject(s)
Hyperoxaluria , Kidney Calculi , Animals , Humans , Hyperoxaluria/complications , Hyperoxaluria/drug therapy , Male , Oxidative Stress , Quercetin/pharmacology , Quercetin/therapeutic use , Rats , Rats, Wistar
3.
Curr Opin Nephrol Hypertens ; 29(4): 400-406, 2020 07.
Article in English | MEDLINE | ID: mdl-32398610

ABSTRACT

PURPOSE OF REVIEW: The aim of the article is to review studies on bone health and oxalate metabolism/therapeutics in the obese rodent model of Roux-en-Y gastric bypass (RYGB) and examine pathways to decrease procedural morbidity. RECENT FINDINGS: Compared with controls, RYGB rodents have up to 40-fold more fat in their stool (steatorrhea) which positively correlates to increased urinary oxalate. These unabsorbed intestinal fatty acids bind calcium and prevent gut calcium oxalate formation, increasing soluble luminal oxalate availability and absorption (enteric hyperoxaluria). When intraluminal fecal fat exceeded about 175 mg/24 h in our model, more paracellular and transcellular oxalate transport across the distal colon occurred. Increasing dietary calcium and colonization with Oxalobacter formigenes reduced hyperoxaluria, whereas vitamin B6 supplementation did not. RYGB animals, when severely calcium deficient, had bone mineral density loss that could not be rescued with vitamin D supplementation. SUMMARY: The findings of hyperoxaluria, steatorrhea, and decreased bone mineral density are seen in both human and rodent RYGB. Our model suggests that a low-fat, low-oxalate diet combined with calcium supplementation can decrease urinary oxalate and improve skeletal bone health. Our model is a useful tool to study renal and bone RYGB effects. Studies of longer duration are required to further evaluate mechanisms of disease and durability of therapeutics.


Subject(s)
Disease Models, Animal , Gastric Bypass , Hyperoxaluria/metabolism , Animals , Bone Density , Humans , Hyperoxaluria/drug therapy , Hyperoxaluria/etiology , Mice , Rats , Steatorrhea/etiology , Steatorrhea/metabolism
4.
Curr Opin Nephrol Hypertens ; 29(4): 394-399, 2020 07.
Article in English | MEDLINE | ID: mdl-32452916

ABSTRACT

PURPOSE OF REVIEW: Oxalate is a metabolic end-product promoting the formation of calcium oxalate crystals in urine. Massive urine oxalate excretion occurs in genetic diseases, mainly primary hyperoxaluria type I and II, threatening renal function. Ethylene glycol poisoning may induce the precipitation of calcium oxalate crystals in renal tubules, leading to acute renal failure. In both cases, oxalate results from glyoxylate transformation to oxalate in the liver, by lactate dehydrogenase (LDH) enzymes, especially the LDH-5 isoenzyme. The purpose of the review is to highlight LDH as a potential therapeutic target according to recent publications. RECENT FINDINGS: Genetic therapy targeting LDH metabolism decreases urine oxalate excretion in rodents. Stiripentol is an antiepileptic drug that has been shown recently to inhibit neuronal LDH-5 isoenzyme. Stiripentol was hypothesized to reduce hepatic oxalate production and urine oxalate excretion. In vitro, stiripentol decreases oxalate synthesis by hepatocytes. In vivo, stiripentol oral administration decreases urine oxalate excretion in rats and protects renal function and renal tissue against ethylene glycol intoxication and chronic calcium oxalate crystalline nephropathy. SUMMARY: The use of stiripentol in-vitro and in-vivo highlights that targeting hepatic LDH by pharmacological or genetic tools may decrease oxalate synthesis, deserving clinical studies.


Subject(s)
Dioxolanes/pharmacology , Hyperoxaluria/drug therapy , Hyperoxaluria/metabolism , Oxalic Acid/metabolism , Humans , Hyperoxaluria, Primary/drug therapy , Hyperoxaluria, Primary/metabolism , L-Lactate Dehydrogenase/metabolism
5.
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
7.
Ren Fail ; 39(1): 193-202, 2017 11.
Article in English | MEDLINE | ID: mdl-27845599

ABSTRACT

BACKGROUND: Star fruit (SF) is a popular fruit, commonly cultivated in many tropical countries, that contains large amount of oxalate. Acute oxalate nephropathy and direct renal tubular damage through release of free radicals are the main mechanisms involved in SF-induced acute kidney injury (AKI). The aim of this study was to evaluate the protective effect of N-acetylcysteine (NAC) on SF-induced nephrotoxicity due to its potent antioxidant effect. MATERIALS AND METHODS: Male Wistar rats received SF juice (4 mL/100 g body weight) by gavage after a 12 h fasting and water deprivation. Fasting and water deprivation continued for 6 h thereafter to warrant juice absorption. Thereafter, animals were allocated to three experimental groups: SF (n = 6): received tap water; SF + NAC (n = 6): received NAC (4.8 g/L) in drinking water for 48 h after gavage; and Sham (n = 6): no interventions. After 48 h, inulin clearance studies were performed to determine glomerular filtration rate. In a second series of experiment, rats were housed in metabolic cages for additional assessments. RESULTS: SF rats showed markedly reduced inulin clearance associated with hyperoxaluria, renal tubular damage, increased oxidative stress and inflammation. NAC treatment ameliorated all these alterations. Under polarized light microscopy, SF rats exhibited intense calcium oxalate birefringence crystals deposition, dilation of renal tubules and tubular epithelial degeneration, which were attenuate by NAC therapy. CONCLUSIONS: Our data show that therapeutic NAC attenuates renal dysfunction in a model of acute oxalate nephropathy following SF ingestion by reducing oxidative stress, oxaluria, and inflammation. This might represent a novel indication of NAC for the treatment of SF-induced AKI.


Subject(s)
Acetylcysteine/pharmacology , Acute Kidney Injury/drug therapy , Antioxidants/pharmacology , Averrhoa/adverse effects , Oxidative Stress/drug effects , Protective Agents/pharmacology , Acute Kidney Injury/chemically induced , Animals , Creatinine/metabolism , Fruit/adverse effects , Glomerular Filtration Rate , Hyperoxaluria/drug therapy , Kidney/physiopathology , Male , Oxalates/adverse effects , Rats , Rats, Wistar
8.
Am J Nephrol ; 44(2): 150-8, 2016.
Article in English | MEDLINE | ID: mdl-27529510

ABSTRACT

BACKGROUND: Hyperoxaluria may result from increased endogenous production or overabsorption of dietary oxalate in the gastrointestinal tract leading to nephrolithiasis and, in some, to oxalate nephropathy and chronic kidney disease. ALLN-177 is an oral formulation of a recombinant, oxalate specific, microbial enzyme oxalate decarboxylase intended to treat secondary hyperoxaluria by degrading dietary oxalate in the gastrointestinal tract, thereby reducing its absorption and subsequent excretion in the urine. METHODS: This double-blind, placebo controlled, randomized, cross-over, phase 1 study of ALLN-177 evaluated the tolerability of ALLN-177 and its effect on urinary oxalate excretion in 30 healthy volunteers with hyperoxaluria induced by ingestion of a high oxalate, low calcium (HOLC) diet. The primary end point was the difference in the mean 24-hour urinary oxalate excretion during the ALLN-177 treatment period compared with the placebo treatment period. RESULTS: The daily urinary oxalate excretion increased in the study population from 27.2 ± 9.5 mg/day during screening to 80.8 ± 24.1 mg/day (mean ± SD) on the HOLC diet before introducing ALLN-177 or placebo therapy for 7 days. Compared to placebo, ALLN-177 treatment reduced urinary oxalate by 11.6 ± 2.7 mg/day, p = 0.0002 (least squares mean ± SD). CONCLUSIONS: In healthy volunteers, with diet-induced hyperoxaluria treatment with ALLN-177, when compared to placebo, significantly reduced urinary oxalate excretion by degrading dietary oxalate in the gastrointestinal tract and thereby reducing its absorption. ALLN-177 may represent a new approach for managing secondary hyperoxaluria and its complications.


Subject(s)
Bacillus subtilis/enzymology , Bacterial Proteins/therapeutic use , Carboxy-Lyases/therapeutic use , Hyperoxaluria/drug therapy , Kidney Calculi/prevention & control , Oxalates/metabolism , Administration, Oral , Adult , Bacterial Proteins/administration & dosage , Bacterial Proteins/adverse effects , Carboxy-Lyases/administration & dosage , Carboxy-Lyases/adverse effects , Cross-Over Studies , Diet/adverse effects , Double-Blind Method , Female , Gastrointestinal Absorption/drug effects , Healthy Volunteers , Humans , Hyperoxaluria/chemically induced , Hyperoxaluria/urine , Male , Middle Aged , Oxalates/pharmacology , Oxalates/urine , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Renal Elimination
9.
Int J Urol ; 21(7): 707-13, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24661223

ABSTRACT

OBJECTIVES: To experimentally evaluate the clinical application of N-methyl-4-isoleucine cyclosporin, a novel selective inhibitor of cyclophilin D activation. METHODS: In vitro, cultured renal tubular cells were exposed to calcium oxalate monohydrate crystals and treated with N-methyl-4-isoleucine cyclosporin. The mitochondrial membrane was stained with tetramethylrhodamine ethyl ester perchlorate and observed. In vivo, Sprague-Dawley rats were divided into four groups: a control group, an ethylene glycol group (administration of ethylene glycol to induce renal calcium crystallization), a N-methyl-4-isoleucine cyclosporin group (administration of N-methyl-4-isoleucine cyclosporin) and an ethylene glycol + N-methyl-4-isoleucine cyclosporin group (administration of ethylene glycol and N-methyl-4-isoleucine cyclosporin). Renal calcium crystallization was evaluated using Pizzolato staining. Oxidative stress was evaluated using superoxide dismutase and 8-hydroxy-deoxyguanosine. Mitochondria within renal tubular cells were observed by transmission electron microscopy. Cell apoptosis was evaluated using cleaved caspase-3. RESULTS: In vitro, calcium oxalate monohydrate crystals induced depolarization of the mitochondrial membrane potential, which was remarkably prevented by N-methyl-4-isoleucine cyclosporin. In vivo, ethylene glycol administration induced renal calcium crystallization, oxidative stress, mitochondrial collapse and cell apoptosis in rats, which were significantly prevented by N-methyl-4-isoleucine cyclosporin. CONCLUSIONS: Herein we first report a new treatment agent determining renal calcium crystallization through cyclophilin D activation.


Subject(s)
Calcium Oxalate/chemistry , Cyclophilins/antagonists & inhibitors , Cyclosporine/pharmacology , Hyperoxaluria/drug therapy , Mitochondria/drug effects , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Calcium Oxalate/pharmacology , Cell Line , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Cyclosporine/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hyperoxaluria/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/ultrastructure , Male , Membrane Potential, Mitochondrial/drug effects , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/ultrastructure , Rats, Sprague-Dawley
10.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 36(3): 313-6, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24997827

ABSTRACT

OBJECTIVE: To observe the therapeutic effect of inulin on enteric hyperoxaluria in rats. METHODS: In experimental A, 24 healthy male Sprague-Dawley rats received an oxalate-free diet on day 1, a high-oxalate diet (oxalate, 74.82 mg/100 g feed stuffs) on days 2 and 3, and plus 2 g inulin to each rat on day 3. The 24-hour urinary volume, concentrations of urinary oxalate and urine creatinine were measured, and 24-hour urinary oxalate excretion was calculated. In experimental B, 24 healthy male Sprague-Dawley rats were equally randomized into control group and inulin group, Each rat received a high oxalate diet (oxalate, 74.82 mg/100 g feedstuffs), and plus 2 g inulin in inulin group. The 24-hour urinary oxalate excretion was calculated in both two groups. RESULTS: In experimental A, the 24-hour urinary oxalate excretion varied with time (F=11.481, P=0.035). The 24-hour urinary oxalate excretion significantly increased on day 2 compared with that on day 1 (P=0.026) and day 3 (P=0.037); it significantly increased on day 3 compared with day 1 (P=0.004). In experimental B, the 24-hour urinary oxalate excretion significantly decreased in inulin group compared with the control (P=0.011). CONCLUSION: Inulin may have potential therapeutic effect on enteric hyperoxaluria in rats.


Subject(s)
Hyperoxaluria/drug therapy , Inulin/therapeutic use , Animals , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Treatment Outcome
11.
Klin Med (Mosk) ; 91(8): 49-53, 2013.
Article in Russian | MEDLINE | ID: mdl-24437186

ABSTRACT

This work was aimed at estimating the role of special therapy of patients with bronchial asthma, COPD and hyperoxaluria designed to restrict the delivery or eliminate excess of oxalates and producing well apparent beneficial effect confirmed by the disappearance or decrease of clinical and functional manifestations of obstruction, reduction of requirements for broncholytic and anti-inflammatory agents.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Asthma , Bronchodilator Agents/therapeutic use , Hyperoxaluria , Oxalic Acid/metabolism , Pulmonary Disease, Chronic Obstructive , Asthma/complications , Asthma/drug therapy , Asthma/metabolism , Humans , Hyperoxaluria/complications , Hyperoxaluria/drug therapy , Hyperoxaluria/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/therapy , Treatment Outcome
12.
Nat Rev Nephrol ; 19(2): 123-138, 2023 02.
Article in English | MEDLINE | ID: mdl-36329260

ABSTRACT

Oxalate homeostasis is maintained through a delicate balance between endogenous sources, exogenous supply and excretion from the body. Novel studies have shed light on the essential roles of metabolic pathways, the microbiome, epithelial oxalate transporters, and adequate oxalate excretion to maintain oxalate homeostasis. In patients with primary or secondary hyperoxaluria, nephrolithiasis, acute or chronic oxalate nephropathy, or chronic kidney disease irrespective of aetiology, one or more of these elements are disrupted. The consequent impairment in oxalate homeostasis can trigger localized and systemic inflammation, progressive kidney disease and cardiovascular complications, including sudden cardiac death. Although kidney replacement therapy is the standard method for controlling elevated plasma oxalate concentrations in patients with kidney failure requiring dialysis, more research is needed to define effective elimination strategies at earlier stages of kidney disease. Beyond well-known interventions (such as dietary modifications), novel therapeutics (such as small interfering RNA gene silencers, recombinant oxalate-degrading enzymes and oxalate-degrading bacterial strains) hold promise to improve the outlook of patients with oxalate-related diseases. In addition, experimental evidence suggests that anti-inflammatory medications might represent another approach to mitigating or resolving oxalate-induced conditions.


Subject(s)
Hyperoxaluria , Renal Insufficiency, Chronic , Renal Insufficiency , Humans , Oxalates/metabolism , Oxalates/pharmacology , Oxalates/therapeutic use , Renal Dialysis , Kidney/metabolism , Hyperoxaluria/therapy , Hyperoxaluria/drug therapy , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency/complications , Homeostasis
13.
Commun Biol ; 6(1): 270, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36922584

ABSTRACT

Hyperoxaluria is well known to cause renal injury and end-stage kidney disease. Previous studies suggested that acetate treatment may improve the renal function in hyperoxaluria rat model. However, its underlying mechanisms remain largely unknown. Using an ethylene glycol (EG)-induced hyperoxaluria rat model, we find the oral administration of 5% acetate reduced the elevated serum creatinine, urea, and protected against hyperoxaluria-induced renal injury and fibrosis with less infiltrated macrophages in the kidney. Treatment of acetate in renal tubular epithelial cells in vitro decrease the macrophages recruitment which might have reduced the oxalate-induced renal tubular cells injury. Mechanism dissection suggests that acetate enhanced acetylation of Histone H3 in renal tubular cells and promoted expression of miR-493-3p by increasing H3K9 and H3K27 acetylation at its promoter region. The miR-493-3p can suppress the expression of macrophage migration inhibitory factor (MIF), thus inhibiting the macrophages recruitment and reduced oxalate-induced renal tubular cells injury. Importantly, results from the in vivo rat model also demonstrate that the effects of acetate against renal injury were weakened after blocking the miR-493-3p by antagomir treatment. Together, these results suggest that acetate treatment ameliorates the hyperoxaluria-induced renal injury via inhibiting macrophages infiltration with change of the miR-493-3p/MIF signals. Acetate could be a new therapeutic approach for the treatment of oxalate nephropathy.


Subject(s)
Acetates , Hyperoxaluria , Macrophage Migration-Inhibitory Factors , MicroRNAs , Animals , Rats , Acetates/pharmacology , Hyperoxaluria/complications , Hyperoxaluria/drug therapy , Hyperoxaluria/genetics , Intramolecular Oxidoreductases/metabolism , Kidney/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Oxalates/adverse effects
14.
Cell Cycle ; 22(17): 1884-1899, 2023 09.
Article in English | MEDLINE | ID: mdl-37592762

ABSTRACT

Nephrolithiasis is a common and frequently-occurring disease in the urinary system with high recurrence. The present study aimed to explore the protective effect and underlying mechanism of hydroxycitric acid (HCA) in hyperoxaluria-induced nephrolithiasis in vitro and in vivo. Crystal deposition and pathophysiological injury in rat models of glyoxylate-induced nephrolithiasis were examined using H&E staining. Cell models of nephrolithiasis were established by oxalate-treated renal tubular epithelial cells. The levels of oxidative stress indexes were determined by ELISA kits. Cell proliferation in vivo and in vitro was evaluated using a cell counting kit-8 (CCK-8) assay and Ki-67 cell proliferation detection kit. Cell apoptosis was measured by flow cytometry and TUNEL staining. The protein levels were examined by western blotting. Our results showed that HCA administration significantly reduced crystal deposition and kidney injury induced by glyoxylate. HCA also alleviated oxidative stress via upregulating the antioxidant enzyme activities of superoxide dismutase (SOD) and catalase (CAT) and reducing the malondialdehyde (MDA) content. Moreover, HCA treatment promoted cell proliferation and inhibited apoptosis of renal tubular epithelial cells exposed to hyperoxaluria. Of note, Nrf2 activator dimethyl fumarate (DMF) exerted the same beneficial effects as HCA in nephrolithiasis. Mechanistically, HCA prevented crystal deposition and oxidative stress induced by hyperoxaluria through targeting the Nrf2/Keap1 antioxidant defense pathway, while knockdown of Nrf2 significantly abrogated these effects. Taken together, HCA exhibited antioxidation and anti-apoptosis activities in nephrolithiasis induced by hyperoxaluria via activating Nrf2/Keap1 pathway, suggesting that it may be an effective therapeutic agent for the prevention and treatment of nephrolithiasis.


Subject(s)
Hyperoxaluria , Nephrolithiasis , Rats , Animals , Antioxidants/pharmacology , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Nephrolithiasis/drug therapy , Nephrolithiasis/metabolism , Oxidative Stress , Hyperoxaluria/complications , Hyperoxaluria/drug therapy , Hyperoxaluria/metabolism , Signal Transduction , Glyoxylates/pharmacology , Glyoxylates/therapeutic use
15.
JCI Insight ; 7(13)2022 07 08.
Article in English | MEDLINE | ID: mdl-35608921

ABSTRACT

Nephrolithiasis is a common and recurrent disease affecting 9% of the US population. Hyperoxaluria is major risk factor for calcium oxalate kidney stones, which constitute two-thirds of all kidney stones. SLC26A3 (DRA, downregulated in adenoma) is an anion exchanger of chloride, bicarbonate, and oxalate thought to facilitate intestinal oxalate absorption, as evidenced by approximately 70% reduced urine oxalate excretion in knockout mice. We previously identified a small-molecule SLC26A3 inhibitor (DRAinh-A270) that selectively inhibited SLC26A3-mediated chloride/bicarbonate exchange (IC50 ~ 35 nM) and, as found here, oxalate/chloride exchange (IC50 ~ 60 nM). In colonic closed loops in mice, luminal DRAinh-A270 inhibited oxalate absorption by 70%. Following oral sodium oxalate loading in mice, DRAinh-A270 largely prevented the 2.5-fold increase in urine oxalate/creatinine ratio. In a mouse model of oxalate nephropathy produced by a high-oxalate low-calcium diet, vehicle-treated mice developed marked hyperoxaluria with elevated serum creatinine, renal calcium oxalate crystal deposition, and renal injury, which were largely prevented by DRAinh-A270 (10 mg/kg twice daily). DRAinh-A270 administered over 7 days to healthy mice did not show significant toxicity. Our findings support a major role of SLC26A3 in intestinal oxalate absorption and suggest the therapeutic utility of SLC26A3 inhibition for treatment of hyperoxaluria and prevention of calcium oxalate nephrolithiasis.


Subject(s)
Hyperoxaluria , Kidney Calculi , Animals , Antiporters , Bicarbonates , Calcium Oxalate , Chlorides/metabolism , Hyperoxaluria/drug therapy , Hyperoxaluria/etiology , Kidney Calculi/complications , Kidney Calculi/drug therapy , Kidney Calculi/prevention & control , Mice , Oxalates , Sulfate Transporters
16.
J Food Biochem ; 46(12): e14377, 2022 12.
Article in English | MEDLINE | ID: mdl-35994414

ABSTRACT

It is well proved that hyperoxaluria induces the renal injury and finally causes the end stage kidney disease. Daphnetin (coumarin derivative) already confirmed renal protective effect in renal model, but hyperoxaluria protective effect still unexplore. The objective of this research was to scrutinize the renal protective effect of daphnetin against ethylene glycol (GC)-induced hyperoxaluria via altering the gut microbiota. GC (1% v/v) was used for the induction of hyperoxaluria in the rats and the rats were received the oral administration of daphnetin (5, 10 and 15 mg/kg). The body and renal weight were assessed. Urine, renal, inflammatory cytokines, antioxidant, inflammatory parameters, and gut microbiota were appraised. Daphnetin effectually improved the body weight and reduced the renal weight. Its also remarkably boosted the magnesium, calcium, citrate level and suppressed the level of uric acid and oxalate formation. Daphnetin significantly (p < .001) ameliorate the level of urinary kidney injury molecule 1 (KIM-1), blood urea nitrogen (BUN), urea, serum creatinine (Scr), neutrophil gelatinase-associated lipocalin (NGAL) and uric acid along with inflammatory cytokines and inflammatory mediators. Daphnetin considerably repressed the malonaldehyde (MDA) level, protein carbonyl and improved the level of glutathione reductase (GR), superoxide dismutase (SOD), glutathione (GSH) and catalase (CAT). Daphnetin treatment considerably altered the microbial composition of different bacteria at phylum, genus and family level. Daphnetin significantly suppressed the Firmicutes relative abundance and boosted the Bacteroidetes relative abundance. Our result clearly indicated that daphnetin remarkably ameliorates the GC induced hyperoxaluria in rats via altering the oxidative stress, inflammatory reaction and gut microbiota. PRACTICAL APPLICATION: Nephrotoxicity is a serious health disease worldwide. We induce the renal toxicity in the experimental rats using the ethylene glycol and scrutinized the renal protective effect of daphnetin. Daphnetin considerably suppress the renal, urine parameters. For estimation the underlying mechanism, we estimated the gut microbiota in all group rats. Daphnetin remarkably altered the level of gut microbiota and suggesting the renal protective effect.


Subject(s)
Gastrointestinal Microbiome , Hyperoxaluria , Renal Insufficiency , Rats , Animals , Uric Acid , Kidney/metabolism , Hyperoxaluria/complications , Hyperoxaluria/drug therapy , Hyperoxaluria/chemically induced , Glutathione/metabolism , Cytokines/metabolism , Ethylene Glycols/adverse effects , Ethylene Glycols/metabolism
17.
Kidney Int ; 80(11): 1146-58, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21866092

ABSTRACT

Hyperoxaluria leads to urinary calcium oxalate (CaOx) supersaturation, resulting in the formation and retention of CaOx crystals in renal tissue. CaOx crystals may contribute to the formation of diffuse renal calcifications (nephrocalcinosis) or stones (nephrolithiasis). When the innate renal defense mechanisms are suppressed, injury and progressive inflammation caused by these CaOx crystals, together with secondary complications such as tubular obstruction, may lead to decreased renal function and in severe cases to end-stage renal failure. For decades, research on nephrocalcinosis and nephrolithiasis mainly focused on both the physicochemistry of crystal formation and the cell biology of crystal retention. Although both have been characterized quite well, the mechanisms involved in establishing urinary supersaturation in vivo are insufficiently understood, particularly with respect to oxalate. Therefore, current therapeutic strategies often fail in their compliance or effectiveness, and CaOx stone recurrence is still common. As the etiology of hyperoxaluria is diverse, a good understanding of how oxalate is absorbed and transported throughout the body, together with a better insight in the regulatory mechanisms, is crucial in the setting of future treatment strategies of this disorder. In this review, the currently known mechanisms of oxalate handling in relevant organs will be discussed in relation to the different etiologies of hyperoxaluria. Furthermore, future directions in the treatment of hyperoxaluria will be covered.


Subject(s)
Hyperoxaluria/etiology , Animals , Calcium Oxalate/metabolism , Humans , Hyperoxaluria/drug therapy , Hyperoxaluria/pathology , Intestinal Diseases/etiology , Kidney Diseases/etiology
18.
Kidney Int ; 79(4): 385-92, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20927039

ABSTRACT

Urolithiasis is a worldwide problem with significant health and economic burdens. Medical therapy that alters the course of stone disease has enormous medical and financial impact. Urolithiasis is a final manifestation of a broad range of etiologies and pathogenesis. The modest progress in understanding the pathophysiology has hampered successful development of targeted therapy. Current regimens are based mostly on rational alteration of urinary biochemistry and physical chemistry to lower the risk of precipitation. In terms of pharmacotherapy, there are drugs to successfully improve hypercalciuria, hypocitraturia, aciduria, hyperuricosuria, and hypercystinuria. These agents have been proven to be effective in randomized controlled trials in improving urinary biochemical and physicochemical risk factors, as well as clinical outcomes. Although our current regimens have clearly improved the management and lives of stone formers, there are still clearly identifiable immense voids in the knowledge of pathophysiology of stone disease that can be filled with combined basic science and clinical studies.


Subject(s)
Urolithiasis/drug therapy , Citric Acid/urine , Cystinuria/drug therapy , Humans , Hypercalciuria/drug therapy , Hyperoxaluria/drug therapy , Randomized Controlled Trials as Topic , Urinary Calculi/chemistry , Urolithiasis/urine
19.
Kidney Int ; 79(5): 538-45, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21085110

ABSTRACT

Adhesion of calcium oxalate (CaOx) crystals to kidney cells may be a key event in the pathogenesis of kidney stones associated with marked hyperoxaluria. Previously, we found that 1,2,3,4,6-penta-O-galloyl-ß-D-glucose (PGG), isolated from a traditional medicinal herb, reduced CaOx crystal adhesion to renal epithelial cells by acting on the cells as well as on the crystal surface. Here we used the ethylene glycol (EG)-mediated hyperoxaluric rat model and found evidence of oxidant stress as indicated by decreases in the activities of the renal antioxidant enzymes, superoxide dismutase, catalase, and glutathione peroxidase, with increased kidney cell apoptosis and serum malondialdehyde levels, all evident by 21 days of EG treatment. These effects of hyperoxaluria were reversed by concurrent PGG treatment along with decreased urinary oxalate levels and CaOx supersaturation. Renal epithelial cell expression of the crystal binding molecule hyaluronan increased diffusely within 7 days of EG initiation, suggesting it is not a result of but precedes crystal deposition. Renal cell osteopontin (OPN) was also upregulated in EG-treated animals, and PGG significantly attenuated overexpression of both OPN and hyaluronan. Thus, our findings demonstrate that PGG reduces renal crystallization and oxidative renal cell injury, and may be a candidate chemopreventive agent for nephrolithiasis.


Subject(s)
Hydrolyzable Tannins/therapeutic use , Hyperoxaluria/drug therapy , Kidney Calculi/prevention & control , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Crystallization , Ethylene Glycol , Hyaluronic Acid/analysis , Hydrolyzable Tannins/pharmacology , Kidney/metabolism , Male , Osteopontin/analysis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
20.
Pharm Biol ; 49(12): 1224-33, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21846174

ABSTRACT

INTRODUCTION: Boerhaavia diffusa Linn. (Nyctaginaceae) is widely used in traditional Indian medicines against renal afflictions including calcium oxalate (CaOx) urolithiasis and is known for antioxidant activity. OBJECTIVE: The present study was designed to investigate the ameliorating effect of aqueous extract of B. diffusa roots (BDE) in hyperoxaluric oxidative stress and renal cell injury. MATERIAL AND METHODS: In vitro antioxidant activity of BDE was estimated in terms of total phenolic content and 1,1-diphenyl-2-picryl hydrazyl free radical scavenging activity. Wistar albino rats were given 0.75% v/v ethylene glycol in drinking water to induce chronic hyperoxaluria and simultaneously BDE was given to nephrolithiasic treated rats at the dose of 100 and 200 mg/kg b.w. orally for 28 days. Urinary volume, oxalate, serum creatinine, blood urea nitrogen (BUN), malondialdehyde (MDA) and antioxidant enzyme (SOD, CAT, GST, GPx) were evaluated. RESULTS AND DISCUSSION: BDE extract was found to posses a high total phenolic content and exhibited significant free radicals scavenging activity. Oxalate excretion significantly increased in hyperoxaluric animals as compared to control which was protected in BDE-treated animals. BDE treatment significantly reduced level of MDA and improved the activity of antioxidant enzymes followed by reduction in BUN and serum creatinine. In addition, BDE reduced the number of CaOx monohydrate crystals in the urine. Histological analysis depicted that BDE treatment inhibited deposition of CaOx crystal and renal cell damage. CONCLUSION: The present study reveals that antioxidant activity of BDE significantly protects against hyperoxaluric oxidative stress and renal cell injury in urolithiasis.


Subject(s)
Acute Kidney Injury/drug therapy , Hyperoxaluria/drug therapy , Nyctaginaceae/chemistry , Oxidative Stress/drug effects , Plant Extracts/therapeutic use , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Antioxidants/metabolism , Biphenyl Compounds , Catalase/metabolism , Chromatography, Thin Layer , Ethylene Glycol , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Hyperoxaluria/chemically induced , Kidney/pathology , Kidney Function Tests , Male , Malondialdehyde/metabolism , Oxalic Acid/metabolism , Picrates , Plant Extracts/chemistry , Plant Roots/chemistry , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Water
SELECTION OF CITATIONS
SEARCH DETAIL