Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 316(1): G1-G14, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30307745

ABSTRACT

Most kidney stones (KS) are composed of calcium oxalate and small increases in urine oxalate enhance the stone risk. Obesity is a risk factor for KS, and urinary oxalate excretion increases with increased body size. We previously established the obese ob/ob ( ob) mice as a model (3.3-fold higher urine oxalate) to define the pathogenesis of obesity-associated hyperoxaluria (OAH). The purpose of this study was to test the hypothesis that the obesity-associated enhanced small intestinal paracellular permeability contributes to OAH by increasing passive paracellular intestinal oxalate absorption. ob Mice have significantly higher jejunal (1.6-fold) and ileal (1.4-fold) paracellular oxalate absorption ex vivo and significantly higher (5-fold) urine [13C]oxalate following oral gavage with [13C]oxalate, indicating increased intestinal oxalate absorption in vivo. The observation of higher oxalate absorption in vivo compared with ex vivo suggests the possibility of increased paracellular permeability along the entire gut. Indeed, ob mice have significantly higher fractions of the administered sucrose (1.7-fold), lactulose (4.4-fold), and sucralose (3.1-fold) excreted in the urine, reflecting increased gastric, small intestinal, and colonic paracellular permeability, respectively. The ob mice have significantly reduced gastrointestinal occludin, zonula occludens-1, and claudins-1 and -3 mRNA and total protein expression. Proinflammatory cytokines and oxidative stress, which are elevated in obesity, significantly enhanced paracellular intestinal oxalate absorption in vitro and ex vivo. We conclude that obese mice have significantly higher intestinal oxalate absorption and enhanced gastrointestinal paracellular permeability in vivo, which would likely contribute to the pathogenesis of OAH, since there is a transepithelial oxalate concentration gradient to drive paracellular intestinal oxalate absorption. NEW & NOTEWORTHY This study shows that the obese ob/ob mice have significantly increased gastrointestinal paracellular oxalate absorption and remarkably enhanced paracellular permeability along the entire gut in vivo, which are likely mediated by the obesity-associated increased systemic and intestinal inflammation and oxidative stress. A transepithelial oxalate concentration gradient driving gastrointestinal paracellular oxalate absorption exists, and therefore, our novel findings likely contribute to the hyperoxaluria observed in the ob/ob mice and hence to the pathogenesis of obesity-associated hyperoxaluria.


Subject(s)
Gastrointestinal Tract/metabolism , Hyperoxaluria/physiopathology , Intestinal Mucosa/metabolism , Obesity/metabolism , Animals , Inflammation/metabolism , Intestinal Absorption/physiology , Intestine, Small/metabolism , Jejunum/metabolism , Mice, Inbred C57BL , Permeability
2.
Kidney Int ; 93(5): 1098-1107, 2018 05.
Article in English | MEDLINE | ID: mdl-29395336

ABSTRACT

Most kidney stones are composed of calcium oxalate, and minor changes in urine oxalate affect the stone risk. Obesity is a risk factor for kidney stones and a positive correlation of unknown etiology between increased body size, and elevated urinary oxalate excretion has been reported. Here, we used obese ob/ob (ob) mice to elucidate the pathogenesis of obesity-associated hyperoxaluria. These ob mice have significant hyperoxaluria (3.3-fold) compared with control mice, which is not due to overeating as shown by pair-feeding studies. Dietary oxalate removal greatly ameliorated this hyperoxaluria, confirming that it is largely enteric in origin. Transporter SLC26A6 (A6) plays an essential role in active transcellular intestinal oxalate secretion, and ob mice have significantly reduced jejunal A6 mRNA (- 80%) and total protein (- 62%) expression. While net oxalate secretion was observed in control jejunal tissues mounted in Ussing chambers, net absorption was seen in ob tissues, due to significantly reduced secretion. We hypothesized that the obesity-associated increase in intestinal and systemic inflammation, as reflected by elevated proinflammatory cytokines, suppresses A6-mediated intestinal oxalate secretion and contributes to obesity-associated hyperoxaluria. Indeed, proinflammatory cytokines (elevated in ob mice) significantly decreased intestinal oxalate transport in vitro by reducing A6 mRNA and total protein expression. Proinflammatory cytokines also significantly reduced active mouse jejunal oxalate secretion, converting oxalate transport from net secretion in vehicle-treated tissues to net absorption in proinflammatory cytokines-treated tissues. Thus, reduced active intestinal oxalate secretion, likely secondary to local and systemic inflammation, contributes to the pathogenesis of obesity-associated hyperoxaluria. Hence, proinflammatory cytokines represent potential therapeutic targets.


Subject(s)
Hyperoxaluria/etiology , Intestinal Secretions/metabolism , Jejunum/metabolism , Obesity/complications , Oxalates/metabolism , Animals , Antiporters/metabolism , Caco-2 Cells , Cytokines/metabolism , Disease Models, Animal , Down-Regulation , Humans , Hyperoxaluria/metabolism , Hyperoxaluria/physiopathology , Inflammation Mediators/metabolism , Intestinal Absorption , Jejunum/physiopathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Obesity/metabolism , Obesity/physiopathology , Secretory Pathway , Sulfate Transporters/metabolism
3.
Toxicol Mech Methods ; 28(3): 195-204, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28980857

ABSTRACT

Experimental induction of hyperoxaluria by ethylene glycol (EG) administration is disapproved as it causes metabolic acidosis while the oral administration of chemically synthesized potassium oxalate (KOx) diet does not mimic our natural system. Since existing models comprise limitations, this study is aimed to develop an improved model for the induction of dietary hyperoxaluria, and nephrocalcinosis in experimental rats by administration of naturally available oxalate rich diet. Male albino Wistar rats were divided into five groups. Group I, control; group II rats received 0.75% EG, group III rats fed with 5% KOx diet and group IV and V rats were administered with spinach extract of 250 and 500 mg soluble oxalate/day respectively, for 28 d. Urine and serum biochemistry were analyzed. After the experimental period, rats were sacrificed, liver and kidney tissue homogenates were used for antioxidant and lipid peroxidation assay. Relative change in expression of kidney injury molecule-1 (KIM-1) and crystal modulators genes in kidney tissues were evaluated. Tissue damage was assessed by histology studies of liver and kidney. Experimental group rats developed hyperoxaluria and crystalluria. Urine parameters, serum biochemistry, antioxidant profile, lipid peroxidation levels and gene expression analysis of experimental group II and III rats reflected acute kidney damage compared to group V rats. Histopathology results showed moderate hyperplasia in liver and severe interstitial inflammation in kidneys of group II and III than group V rats. Ingestion of naturally available oxalate enriched spinach extract successfully induced dietary hyperoxaluria and nephrocalcinosis in rats with minimal kidney damage.


Subject(s)
Disease Models, Animal , Foodborne Diseases/etiology , Hyperoxaluria/etiology , Nephrocalcinosis/etiology , Oxalic Acid/poisoning , Plant Leaves/adverse effects , Spinacia oleracea/adverse effects , Administration, Oral , Animals , Biomarkers/blood , Biomarkers/metabolism , Biomarkers/urine , Crystallization , Ethylene Glycol/toxicity , Foodborne Diseases/metabolism , Foodborne Diseases/pathology , Foodborne Diseases/physiopathology , Gene Expression Regulation/drug effects , Hyperoxaluria/metabolism , Hyperoxaluria/pathology , Hyperoxaluria/physiopathology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Nephrocalcinosis/metabolism , Nephrocalcinosis/pathology , Nephrocalcinosis/physiopathology , Oxalic Acid/administration & dosage , Oxalic Acid/chemistry , Oxalic Acid/metabolism , Plant Extracts/adverse effects , Plant Extracts/chemistry , Plant Leaves/chemistry , Rats, Wistar , Renal Insufficiency/etiology , Spinacia oleracea/chemistry
4.
Pediatr Nephrol ; 32(5): 781-790, 2017 05.
Article in English | MEDLINE | ID: mdl-27924398

ABSTRACT

BACKGROUND: Primary hyperoxaluria (PH) is a rare, genetic disorder which involves the overproduction of endogenous oxalate, leading to hyperoxaluria, recurrent urolithiasis and/or progressive nephrocalcinosis and eventually resulting in kidney failure and systemic oxalosis. The aim of this trial was to investigate whether treatment involving an oxalate-metabolising bacterium (Oxalobacter formigenes) could reduce urinary oxalate excretion in PH patients. METHODS: The efficacy and safety of O. formigenes (Oxabact® OC5; OxThera AB, Stockholm, Sweden) was evaluated in a randomised, placebo-controlled, double-blind study for 8 weeks. The primary objective was reduction in urinary oxalate excretion (Uox). Secondary objectives included faecal O. formigenes count and decrease in plasma oxalate concentration (Pox). RESULTS: Twenty-eight patients randomised 1:1 to the treatment group (OC5) or the placebo group completed the study. After 8 weeks of treatment, there was no significant difference in the change in Uox (mmol/24 h/1.73 m2) between the groups (OC5: +0.042, placebo: -0.140). Post-hoc analysis showed a statistically significant increase in Uox per urinary creatinine excretion in the OC5 group (OC5: +5.41, placebo: -15.96; p = 0.030). Change in Pox from baseline was not significantly different between groups (p = 0.438). The O. formigenes cell count was significantly increased in OC5-treated patients (p < 0.001) versus placebo. The treatment response to O. formigenes was related to individual stage of kidney deterioration, and Pox was directly correlated to kidney function, even for early-stage patients (chronic kidney disease stage 1). No safety issues were observed. CONCLUSIONS: Treatment with OC5 did not significantly reduce Uox or Pox over 8 weeks of treatment. The treatment was well tolerated and successfully delivered to the gastrointestinal tract.


Subject(s)
Hyperoxaluria/therapy , Oxalobacter formigenes , Adolescent , Bacterial Load , Child , Child, Preschool , Double-Blind Method , Feces/microbiology , Female , Humans , Hyperoxaluria/physiopathology , Hyperoxaluria/urine , Kidney Function Tests , Male , Oxalic Acid/urine , Probiotics/administration & dosage , Probiotics/adverse effects , Probiotics/therapeutic use , Tablets, Enteric-Coated , Treatment Outcome , Young Adult
5.
Klin Med (Mosk) ; 94(6): 467-9, 2016.
Article in Russian | MEDLINE | ID: mdl-30289667

ABSTRACT

Objective: This case demonstrates the difficulty of diagnostics of oxalate nephropathy and possibility of development of acute kidney injury. Summary: The paper describes a patient with oxalate nephropathy and acute kidney injury. Specific features of oxalate diathesis are discussed as well as approaches to its diagnosis. Clinical peculiarities and diagnostic difficulties are described. Conclusions: Early diagnosis of oxalate nephropathy and treatment permits to improve prognosis.


Subject(s)
Acute Kidney Injury , Hyperoxaluria , Kidney/diagnostic imaging , Oxalic Acid/metabolism , Pyelonephritis/diagnosis , Renal Dialysis/methods , Acute Kidney Injury/diagnosis , Acute Kidney Injury/etiology , Acute Kidney Injury/physiopathology , Acute Kidney Injury/therapy , Adult , Diagnosis, Differential , Early Diagnosis , Female , Humans , Hyperoxaluria/complications , Hyperoxaluria/diagnosis , Hyperoxaluria/physiopathology , Hyperoxaluria/therapy , Kidney Function Tests/methods , Tomography, X-Ray Computed/methods , Treatment Outcome , Ultrasonography/methods
6.
Mol Cell Biochem ; 405(1-2): 105-14, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25842190

ABSTRACT

The purpose of the present study was to evaluate the nephro-protective potential of N-acetylcysteine against hyperoxaluria-induced renal mitochondrial dysfunction in rats. Nine days dosing of 0.4 % ethylene glycol +1 % ammonium chloride, developed hyperoxaluria in male wistar rats which resulted in renal injury and dysfunction as supported by increased level of urinary lactate dehydrogenase, calcium, and decreased creatinine clearance. Mitochondrial oxidative strain in hyperoxaluric animals was evident by decreased levels of superoxide dismutase, glutathione peroxidase, glutathione reductase, reduced glutathione, and an increased lipid peroxidation. Declined activities of respiratory chain enzymes and tricarboxylic acid cycle enzymes showed mitochondrial dysfunction in hyperoxaluric animals. N-acetylcysteine (50 mg/kg, i.p.), by virtue of its -SH reviving power, was able to increase the glutathione levels and thus decrease the oxidative stress in renal mitochondria. Hence, mitochondrial damage is, evidently, an essential event in ethylene glycol-induced hyperoxaluria and N-acetylcysteine presented itself as a safe and effective remedy in combating nephrolithiasis.


Subject(s)
Acetylcysteine/pharmacology , Hyperoxaluria/physiopathology , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Protective Agents/pharmacology , Animals , Calcium/urine , Citric Acid Cycle/drug effects , Creatinine/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Hyperoxaluria/metabolism , Hyperoxaluria/urine , Kidney/metabolism , Kidney/physiopathology , L-Lactate Dehydrogenase/urine , Lipid Peroxidation/drug effects , Male , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/physiopathology , Mitochondrial Diseases/urine , Nephrolithiasis/metabolism , Nephrolithiasis/physiopathology , Nephrolithiasis/urine , Oxidative Stress/drug effects , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
7.
Exp Physiol ; 99(9): 1163-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24951497

ABSTRACT

Studies have shown that compensatory adaptations in gastrointestinal oxalate transport can impact the amount of oxalate excreted by the kidney. Hyperoxaluria is a major risk factor in the formation of kidney stones, and oxalate is derived from both the diet and the liver metabolism of glyoxylate. Although the intestine generally absorbs oxalate from dietary sources and can contribute as much as 50% of urinary oxalate, enteric oxalate elimination plays a significant role when renal function is compromised. While the mechanistic basis for these changes in the direction of intestinal oxalate movements in chronic renal failure involves an upregulation of angiotensin II receptors in the large intestine, enteric secretion/excretion of oxalate can also occur by mechanisms that are independent of angiotensin II. Most notably, the commensal bacterium Oxalobacter sp. interacts with the host enterocyte and promotes the movement of oxalate from the blood into the lumen, resulting in the beneficial effect of significantly lowering urinary oxalate excretion. Changes in the passive permeability of the intestine, such as in steatorrhoea and following gastric bypass, also promote oxalate absorption and hyperoxaluria. In summary, this report highlights the two-way physiological signalling between the gut and the kidney, which may help to alleviate the consequences of certain kidney diseases.


Subject(s)
Gastric Bypass , Intestinal Mucosa/metabolism , Kidney/metabolism , Oxalic Acid/metabolism , Renal Insufficiency, Chronic/metabolism , Adaptation, Physiological , Animals , Humans , Hyperoxaluria/metabolism , Hyperoxaluria/physiopathology , Intestinal Absorption , Intestinal Elimination , Intestines/microbiology , Intestines/physiopathology , Kidney/physiopathology , Kidney Calculi/metabolism , Kidney Calculi/physiopathology , Oxalic Acid/urine , Permeability , Renal Elimination , Renal Insufficiency, Chronic/physiopathology , Signal Transduction
9.
J Urol ; 190(3): 1102-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23499748

ABSTRACT

PURPOSE: We determined the effect of dietary fat and oxalate on fecal fat excretion and urine parameters in a rat model of Roux-en-Y gastric bypass surgery. MATERIALS AND METHODS: Diet induced obese Sprague-Dawley® rats underwent sham surgery as controls (16), or Roux-en-Y gastric bypass surgery (19). After recovery, rats had free access to a normal calcium, high fat (40%) diet with or without 1.5% potassium oxalate for 5 weeks and then a normal (10%) fat diet for 2 weeks. Stool and urine were collected after each period. Fecal fat was determined by gas chromatography and urine metabolites were evaluated by assay spectrophotometry. RESULTS: Daily fecal fat excretion remained low in controls on either diet. However, Roux-en-Y gastric bypass rats ingested a food quantity similar to that of controls but had eightfold higher fecal fat excretion (p <0.001) and heavier stools (p = 0.02). Compared to controls, gastric bypass rats on the high fat diet with potassium oxalate had a fivefold increase in urine oxalate excretion (p <0.001), while gastric bypass rats without potassium oxalate had a twofold increase in urine calcium (p <0.01). Lowering dietary fat in gastric bypass rats with potassium oxalate led to a 50% decrease in oxalate excretion (p <0.01), a 30% decrease in urine calcium and a 0.3 U increase in urine pH (p <0.001). CONCLUSIONS: In this Roux-en-Y gastric bypass model high fat feeding resulted in steatorrhea, hyperoxaluria and low urine pH, which were partially reversible by lowering the dietary fat and oxalate content. Roux-en-Y gastric bypass rats on normal fat and no oxalate diets excreted twice as much oxalate as age matched, sham operated controls. Although Roux-en-Y gastric bypass hyperoxaluria appears primarily mediated by gut and diet, secondary causes of oxalogenesis from liver or other mechanisms deserve further exploration.


Subject(s)
Dietary Fats/metabolism , Gastric Bypass/adverse effects , Hyperoxaluria/etiology , Obesity/surgery , Oxalates/metabolism , Steatorrhea/etiology , Animals , Disease Models, Animal , Feces/chemistry , Gastric Bypass/methods , Hyperoxaluria/physiopathology , Male , Obesity/complications , Postoperative Complications/epidemiology , Postoperative Complications/physiopathology , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values , Risk Assessment , Steatorrhea/physiopathology , Treatment Outcome , Urinalysis
10.
Life Sci ; 291: 120258, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-34952043

ABSTRACT

AIMS: Enhancer of zeste homolog 2 (EZH2), a histone H3 lysine 27 methyltransferase, has been shown to play a role in kidney diseases. However, its role in hyperoxaluria-induced renal tubular epithelial cells (TECs) injury remains unclear. MATERIALS AND METHODS: A hyperoxaluria rat model was established by providing 0.5% ammonium chloride and drinking water containing 1% ethylene glycol. TECs were exposed to oxalate stress. The 3-DZNeP, a selective EZH2 inhibitor, was administered in vivo and in vitro. Cell viability, ROS production, and apoptosis ratio were evaluated. Crystal deposition was detected by Von Kossa staining and kidney tissue injury was detected by HE staining and TUNEL. EZH2, H3K27me3, cleaved-caspase3, IL-6, and MCP-1 were examined by western blot or immunohistochemistry. KEY FINDINGS: Inhibition of EZH2 by 3-DZNeP significantly attenuated hyperoxaluria-induced oxidative and inflammatory injury and CaOx crystal deposition in vivo. Similarly, inhibition of EZH2 using 3-DZNeP or shRNA restored cell viability, suppressed LDH release and the production of intracellular ROS in vitro. Furthermore, the MAPK signaling pathway and FoxO3a levels were activated or elevated in TECs exposed to oxalate. EZH2 inhibition using 3-DZNeP blocked these effects. CC90003 (ERK inhibitor) or SB203580 (p38 inhibitor) did not significantly affect the expression of FoxO3a in TECs treated with 3-DZNeP and oxalate; only SP600125 (JNK inhibitor) significantly decreased FoxO3a expression. SIGNIFICANCE: EZH2 inhibition protects against oxalate-induced TECs injury and reduces CaOx crystal deposition in the kidney may by modulating the JNK/FoxO3a pathway; EZH2 may be a promising therapeutic target in TECs injury.


Subject(s)
Acute Kidney Injury/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Hyperoxaluria/metabolism , Acute Kidney Injury/physiopathology , Animals , Apoptosis/drug effects , China , Enhancer of Zeste Homolog 2 Protein/physiology , Epithelial Cells/metabolism , Forkhead Box Protein O3/physiology , Hyperoxaluria/physiopathology , Kidney/metabolism , Kidney Diseases/metabolism , MAP Kinase Signaling System/physiology , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
11.
Clin J Am Soc Nephrol ; 16(3): 487-495, 2021 03 08.
Article in English | MEDLINE | ID: mdl-32900691

ABSTRACT

Enteric hyperoxaluria is a distinct entity that can occur as a result of a diverse set of gastrointestinal disorders that promote fat malabsorption. This, in turn, leads to excess absorption of dietary oxalate and increased urinary oxalate excretion. Hyperoxaluria increases the risk of kidney stones and, in more severe cases, CKD and even kidney failure. The prevalence of enteric hyperoxaluria has increased over recent decades, largely because of the increased use of malabsorptive bariatric surgical procedures for medically complicated obesity. This systematic review of enteric hyperoxaluria was completed as part of a Kidney Health Initiative-sponsored project to describe enteric hyperoxaluria pathophysiology, causes, outcomes, and therapies. Current therapeutic options are limited to correcting the underlying gastrointestinal disorder, intensive dietary modifications, and use of calcium salts to bind oxalate in the gut. Evidence for the effect of these treatments on clinically significant outcomes, including kidney stone events or CKD, is currently lacking. Thus, further research is needed to better define the precise factors that influence risk of adverse outcomes, the long-term efficacy of available treatment strategies, and to develop new therapeutic approaches.


Subject(s)
Hyperoxaluria/physiopathology , Hyperoxaluria/therapy , Gastrointestinal Diseases/complications , Humans , Hyperoxaluria/etiology
12.
Neth J Med ; 77(8): 287-292, 2019 10.
Article in English | MEDLINE | ID: mdl-31814577

ABSTRACT

BACKGROUND: In this article, we present two cases of patients with acute renal insufficiency with a history of exocrine pancreatic insufficiency. In one case, this was caused by pancreaticoduodenectomy; in the other, by alcohol abuse. Neither patient had considerable proteinuria or haematuria. Their renal biopsies showed tubulopathy with widespread oxalate crystals, characterised by their birefringence in light microscopy. Restricting oxalate intake and prescribing oxalate binding agents reduced serum oxalate levels. Renal function partially recovered in both patients. Oxalate nephropathy is associated with exocrine pancreatic insufficiency, gastric and pancreatic surgery, and inflammatory bowel disease. Normally, dietary calcium binds oxalate to form calcium oxalate, which is excreted in the stool. In patients with pancreatic insufficiency, fatty acids bind calcium instead, allowing oxalate to be absorbed in the colon. The resulting hyperoxaluria can cause oxalate crystal formation, tubulopathy, and renal insufficiency. Treatment relies on decreasing the amount of absorbable oxalate in the intestinal lumen, as well as lowering urinary oxalate concentrations. CONCLUSION: Secondary hyperoxaluria is a common cause of renal insufficiency and should be considered in patients with a medical history of pancreatic insufficiency and progressive kidney injury.


Subject(s)
Exocrine Pancreatic Insufficiency/complications , Hyperoxaluria/complications , Aged , Alcoholism/complications , Humans , Hyperoxaluria/diagnosis , Hyperoxaluria/physiopathology , Hyperoxaluria/therapy , Male , Middle Aged
13.
Expert Opin Ther Targets ; 23(5): 379-391, 2019 05.
Article in English | MEDLINE | ID: mdl-30905219

ABSTRACT

INTRODUCTION: Oxalate is a toxic byproduct of metabolism and is normally produced in quantities easily removed from the body. However, under specific circumstances oxalate production is increased resulting in deposition of calcium oxalate (CaOx) crystals in the kidneys as well as other organs causing inflammation and injury. Excessive buildup of crystal deposits in the kidneys causes eventual loss of renal function requiring renal transplantation. Areas covered: Cellular exposure to CaOx crystals induces the production of reactive oxygen species (ROS) with the involvement of renin-angiotensin aldosterone system (RAAS), mitochondria, and NADPH oxidase. Inflammasomes are activated and pro-inflammatory cytokines, such as IL-1ß and IL-18 are produced. We reviewed results of experimental and clinical studies of crystal renal epithelial cell interactions with emphasis on cellular injury and ROS production. Expert opinion: Treatment should depend upon the level of hyperoxaluria and whether it is associated with CaOx crystal deposition. Persistent low grade or intermittent hyperoxaluria can be treated with antioxidants, free radical scavengers. Hyperoxaluria associated with CaOx crystal deposition will require administration of angiotensin II receptor blockers, and NADPH oxidase or NLRP3 inflammasome inhibitors. DASH-style diet will be beneficial in both cases.


Subject(s)
Hyperoxaluria/therapy , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Antioxidants/pharmacology , Calcium Oxalate/metabolism , Free Radical Scavengers/pharmacology , Humans , Hyperoxaluria/physiopathology
14.
Am J Med ; 90(4): 498-504, 1991 Apr.
Article in English | MEDLINE | ID: mdl-2012090

ABSTRACT

A 23-year-old man with type 1 primary hyperoxaluria, renal failure, and oxalosis developed a severe cardiomyopathy while awaiting combined liver-kidney transplantation. This manifested as radiographic cardiomegaly, a dilated hypokinetic left ventricle with a decreased ejection fraction, ventricular arrhythmias, and cardiac uptake on bone scanning. On liver and kidney transplantation, these abnormalities markedly improved and/or reversed. The cardiac size almost normalized, the left ventricular ejection fraction increased from 20% to 34%, the ventricular arrhythmias resolved, and the cardiac uptake on bone scanning disappeared. This coincided with normalization of oxalate production and excretion. Severe cardiac involvement secondary to oxalosis in patients with primary hyperoxaluria may improve or reverse with combined liver-kidney transplantation.


Subject(s)
Cardiomyopathies/complications , Hyperoxaluria/complications , Kidney Failure, Chronic/complications , Kidney Transplantation , Liver Transplantation , Adult , Cardiomyopathies/diagnostic imaging , Humans , Hyperoxaluria/physiopathology , Liver/metabolism , Male , Oxalates/metabolism , Postoperative Period , Radiography , Renal Dialysis
15.
Cell Biochem Biophys ; 32 Spring: 171-6, 2000.
Article in English | MEDLINE | ID: mdl-11330044

ABSTRACT

Glyoxylate is an immediate precursor of oxalate, but in its metabolism the conversion into glycine catalyzed by serine:pyruvate/alanine:glyoxylate aminotransferase (SPT/AGT) appears to be the main route. When SPT/AGT is missing as in the case of primary hyperoxaluria type 1 (PH1) more glyoxylate is used for the oxalate production, resulting in calcium oxalate urolithiasis and finally systemic oxalosis. SPT/AGT is a unique enzyme of species-specific dual organelle localization; it is located largely in mitochondria in carnivores and entirely in peroxisomes in herbivores and man. For herbivores, the peroxisomal localization of SPT/AGT is indispensable to avoid massive production of oxalate, probably because liver peroxisomes are the main site of glyoxylate production from glycolate, and plants contain glycolate much more than animal tissues. Recently, we took charge of laboratory examination for 8 cases of primary hyperoxaluria in Japan, and felt that symptoms of some of the Japanese PH1 patients are apparently milder than those of Western patients. The reason of this is not clear, but from the above mentioned seemingly indispensable association of grass-eating with the peroxisomal localization of SPT/AGT it may be related, at least in part, to the food habit of Japanese, especially that of old generation, that they prefer boiled greens rather than frying or raw vegetables.


Subject(s)
Hyperoxaluria/metabolism , Oxalates/metabolism , Diet , Humans , Hyperoxaluria/epidemiology , Hyperoxaluria/physiopathology , Japan/epidemiology , Transaminases/metabolism
16.
Article in English | MEDLINE | ID: mdl-11993719

ABSTRACT

Hyperoxaluria is one of the major risk factors for the formation of urinary calcium oxalate stones. Calcium oxalate crystals and their deposition have been implicated in inducing renal tubular damage. Lipoic acid (LA) and eicosapentaenoic acid (EPA) have been shown to ameliorate the changes associated with hyperoxaluria. This prompted us to investigate the nephroprotectant role of EPA-LA, a new derivative, in vivo in hyperoxaluric rats. Elevation in the levels of calcium, oxalate and phosphorus, the stone-forming constituents, were observed in calculogenic rats as a manifestation of crystal deposition. Tubular damage to the renal tissue was assessed byassaying the excretion of marker enzymes in the urine. Damage to the tubules was indicated by increased excretion of alkaline phosphatase (ALP), lactate dehydrogenase (LDH), gamma-glutamyl transferase (gamma-GT), beta-Glucuronidase (beta-GLU) and N-Acetyl beta-D glucosaminidase (NAG). Fibrinolytic activity was found to be reduced. Administration of EPA, LA and EPA-LA reduced the tubular damage and decreased the markers of crystal deposition markedly, which was substantiated by the reduction in weight of bladder stone formed. Our results highlight that EPA-LA is the most effective drug in inhibiting stone formation and mitigating renal damage caused by oxalate toxicity, thus confirming it as a nephroprotectant. Further work in this direction is warranted to establish the therapeutic effectiveness of this new derivative.


Subject(s)
Eicosapentaenoic Acid/pharmacology , Kidney/drug effects , Oxalates/toxicity , Thioctic Acid/pharmacology , Acetylglucosaminidase/drug effects , Acetylglucosaminidase/metabolism , Animals , Disease Models, Animal , Eicosapentaenoic Acid/analogs & derivatives , Glucuronidase/drug effects , Glucuronidase/metabolism , Hyperoxaluria/drug therapy , Hyperoxaluria/physiopathology , Kidney/metabolism , Kidney/pathology , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase/metabolism , Magnesium/urine , Male , Rats , Rats, Wistar , Thioctic Acid/analogs & derivatives , Urinary Bladder Calculi/drug therapy , gamma-Glutamyltransferase/drug effects , gamma-Glutamyltransferase/metabolism
17.
J Pharm Pharmacol ; 50(5): 501-5, 1998 May.
Article in English | MEDLINE | ID: mdl-9643443

ABSTRACT

This study aimed to evaluate whether administration of cyclosporin to hyperoxaluric rats affects liver antioxidant status, and whether pretreatment with vitamin E reverses the effect. Male Wistar rats were divided into two major groups of 40. One group was given vitamin E. Both major groups were then divided into four subgroups which received vehicle (olive oil), cyclosporin in olive oil (50 mg kg(-1)), 3% ammonium oxalate or cyclosporin + 3% ammonium oxalate for three days. The activities of liver lactate dehydrogenase, glycolic acid oxidase and xanthine oxidase, and the level of malondialdehyde, an indicator of lipid peroxidation, increased when cyclosporin was administered to hyperoxaluric rats. The levels of antioxidants ascorbic acid, vitamin E and reduced glutathione and the activities of glutathione-metabolizing enzymes were altered significantly when hyperoxaluric rats were treated with cyclosporin. All these enzymes and antioxidants showed highly significant correlation values, r. These changes were restored to near normal by pretreatment with vitamin E. These findings suggest that cyclosporin-induced hepatotoxicity is aggravated in hyperoxaluria. This was almost totally prevented by pretreatment with vitamin E.


Subject(s)
Antioxidants/therapeutic use , Cyclosporine/antagonists & inhibitors , Cyclosporine/toxicity , Hyperoxaluria/physiopathology , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/toxicity , Liver/drug effects , Vitamin E/therapeutic use , Animals , Antioxidants/analysis , Antioxidants/pharmacology , Ascorbic Acid/analysis , Drug Interactions , Hyperoxaluria/chemically induced , Lipid Peroxidation/drug effects , Liver/chemistry , Liver/enzymology , Liver Function Tests , Male , Oxalates , Rats , Rats, Wistar , Vitamin E/analysis , Vitamin E/pharmacology
18.
Adv Pediatr ; 47: 199-221, 2000.
Article in English | MEDLINE | ID: mdl-10959444

ABSTRACT

Urinary stones in children are being recognized with increasing frequency. Formerly thought to be the result of dehydration, urinary obstruction, or infection, most urinary calculi in children now are recognized to have an underlying metabolic abnormality. A number of challenges face pediatricians in evaluating and treating children with urinary stone disease. Often the clinical symptomatology is nonspecific and lacks the excruciating renal colic seen in adults. Furthermore, diagnostic clinical laboratory values vary with age and must be differentiated from normal values reported for adult patients. Both environmental and genetic factors are responsible for urinary stones. Many stones have a hereditary basis. Exciting new information is developing about the genetic propensity for urinary stones. Current medical therapies attempt either to reduce the production of a lithogenic solute or to increase urinary solubility. New therapies for prevention and treatment of urinary stone disease are likely to evolve as our understanding of the pathogenesis of these conditions grows.


Subject(s)
Calcium/urine , Urinary Calculi/etiology , Adult , Age Factors , Calcium/metabolism , Calcium Oxalate/urine , Child , Cystinuria/diagnosis , Diagnosis, Differential , Diet/adverse effects , Humans , Hyperoxaluria/diagnosis , Hyperoxaluria/physiopathology , Uric Acid/metabolism , Urinary Calculi/diagnosis , Urinary Calculi/genetics , Urinary Calculi/therapy
19.
Transplant Rev (Orlando) ; 28(4): 182-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24999029

ABSTRACT

Secondary hyperoxaluria is a multifactorial disease affecting several organs and tissues, among which stand native and transplanted kidneys. Nephrocalcinosis and nephrolithiasis may lead to renal insufficiency. Patients suffering from secondary hyperoxaluria, should be promptly identified and appropriately treated, so that less renal damage occurs. The aim of this review is to underline the causes of hyperoxaluria and the related pathophysiologic mechanisms, which are involved, along with the description of seven cases of irreversible renal graft injury due to secondary hyperoxaluria.


Subject(s)
Hyperoxaluria/epidemiology , Renal Insufficiency/epidemiology , Diagnosis, Differential , Disease Progression , Humans , Hyperoxaluria/diagnosis , Hyperoxaluria/etiology , Hyperoxaluria/physiopathology , Kidney Calculi/epidemiology , Kidney Failure, Chronic/pathology , Kidney Transplantation , Malabsorption Syndromes/complications , Risk Factors
20.
Saudi J Kidney Dis Transpl ; 25(1): 113-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24434393

ABSTRACT

Renal oxalate deposition can be seen with primary hyperoxaluria, malabsorptive states, ethylene glycol toxicity and, rarely, with excessive vitamin C ingestion. We report a case of secondary hyperoxaluria in which the diagnosis was not considered initially because there was no past history of urinary calculi and no evidence of nephrocalcinosis on plain X-ray of the abdomen and ultrasonography. The disease was detected and diagnosed only after kidney transplantation. Secondary oxalosis can cause graft loss or delayed graft function. Biopsy of the allograft should be carefully examined for oxalate deposits even in the absence of a family history. When oxalosis is diagnosed, intensifying hemodialysis (HD) to eliminate calcium oxalate can help in the recovery of renal function in some cases. Systematic vitamin C supplementation in HD patients should be avoided as it can be a cause of secondary oxalosis.


Subject(s)
Ascorbic Acid/adverse effects , Hyperoxaluria/chemically induced , Kidney Failure, Chronic/therapy , Kidney Transplantation , Kidney/drug effects , Renal Dialysis , Adult , Biopsy , Female , Humans , Hyperoxaluria/diagnosis , Hyperoxaluria/physiopathology , Hyperoxaluria/therapy , Kidney/pathology , Kidney/physiopathology , Kidney Failure, Chronic/physiopathology , Kidney Failure, Chronic/surgery , Renal Dialysis/adverse effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL