Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Nutr Metab Cardiovasc Dis ; 34(3): 559-568, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38431384

RESUMO

AIMS: Previous studies find kidney stone formers (KSF) are at greater risk of developing cardiovascular disease (CVD). The underlying mechanisms are poorly understood, and many clinicians are unaware of this connection. We will: DATA SYNTHESIS: Our systematic review is registered with PROSPERO (ID CRD42021251477). We searched epidemiological and biological data. The epidemiological search generated 669 papers, narrowed down to 15. There were 4,259,869 participants (230,720 KSFs). KSF was associated with 25% higher risk of coronary artery disease (CAD) (95% confidence interval (CI): 15, 35%), 17% higher risk of stroke/transient ischemic attacks (TIA) (CI:10, 25%) and 39% higher risk of arterial disease (AD) (CI: 17 65%). Significant heterogeneity was found. Female-identifying KSFs had a higher risk of stroke (ratio = 1.10) and CAD (1.20). The biological search generated 125 papers, narrowed down to 14. Potential underlying mechanisms were extracted and discussed, including intimal/medial vascular calcification, oxidative stress via osteopontin (OPN), cholesterol-induced pathology, and endothelial dysfunction. CONCLUSIONS: There is a significant association between KSF and CVD, supporting the consideration of KSF as a systemic, calcium-mediated disease. Clinicians will benefit from being aware of this connection.


Assuntos
Doenças Cardiovasculares , Doença da Artéria Coronariana , Cálculos Renais , Acidente Vascular Cerebral , Humanos , Feminino , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/complicações , Cálculos Renais/diagnóstico , Cálculos Renais/epidemiologia , Cálculos Renais/complicações , Doença da Artéria Coronariana/diagnóstico , Doença da Artéria Coronariana/epidemiologia , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/epidemiologia , Colesterol
2.
Kidney Int Rep ; 8(10): 2029-2042, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37849991

RESUMO

Introduction: Primary hyperoxaluria type 1 (PH1) has a highly heterogeneous disease course. Apart from the c.508G>A (p.Gly170Arg) AGXT variant, which imparts a relatively favorable outcome, little is known about determinants of kidney failure. Identifying these is crucial for disease management, especially in this era of new therapies. Methods: In this retrospective study of 932 patients with PH1 included in the OxalEurope registry, we analyzed genotype-phenotype correlations as well as the impact of nephrocalcinosis, urolithiasis, and urinary oxalate and glycolate excretion on the development of kidney failure, using survival and mixed model analyses. Results: The risk of developing kidney failure was the highest for 175 vitamin-B6 unresponsive ("null") homozygotes and lowest for 155 patients with c.508G>A and c.454T>A (p.Phe152Ile) variants, with a median age of onset of kidney failure of 7.8 and 31.8 years, respectively. Fifty patients with c.731T>C (p.Ile244Thr) homozygote variants had better kidney survival than null homozygotes (P = 0.003). Poor outcomes were found in patients with other potentially vitamin B6-responsive variants. Nephrocalcinosis increased the risk of kidney failure significantly (hazard ratio [HR] 3.17 [2.03-4.94], P < 0.001). Urinary oxalate and glycolate measurements were available in 620 and 579 twenty-four-hour urine collections from 117 and 87 patients, respectively. Urinary oxalate excretion, unlike glycolate, was higher in patients who subsequently developed kidney failure (P = 0.034). However, the 41% intraindividual variation of urinary oxalate resulted in wide confidence intervals. Conclusion: In conclusion, homozygosity for AGXT null variants and nephrocalcinosis were the strongest determinants for kidney failure in PH1.

3.
Kidney Int ; 104(5): 975-984, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37414395

RESUMO

Urinary stone disease (USD) is a major health burden affecting over 10% of the United Kingdom population. While stone disease is associated with lifestyle, genetic factors also strongly contribute. Common genetic variants at multiple loci from genome-wide association studies account for 5% of the estimated 45% heritability of the disorder. Here, we investigated the extent to which rare genetic variation contributes to the unexplained heritability of USD. Among participants of the United Kingdom 100,000-genome project, 374 unrelated individuals were identified and assigned diagnostic codes indicative of USD. Whole genome gene-based rare variant testing and polygenic risk scoring against a control population of 24,930 ancestry-matched controls was performed. We observed (and replicated in an independent dataset) exome-wide significant enrichment of monoallelic rare, predicted damaging variants in the SLC34A3 gene for a sodium-dependent phosphate transporter that were present in 5% cases compared with 1.6% of controls. This gene was previously associated with autosomal recessive disease. The effect on USD risk of having a qualifying SLC34A3 variant was greater than that of a standard deviation increase in polygenic risk derived from GWAS. Addition of the rare qualifying variants in SLC34A3 to a linear model including polygenic score increased the liability-adjusted heritability from 5.1% to 14.2% in the discovery cohort. We conclude that rare variants in SLC34A3 represent an important genetic risk factor for USD, with effect size intermediate between the fully penetrant rare variants linked with Mendelian disorders and common variants associated with USD. Thus, our findings explain some of the heritability unexplained by prior common variant genome-wide association studies.


Assuntos
Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc , Cálculos Urinários , Urolitíase , Doenças Urológicas , Humanos , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Polimorfismo de Nucleotídeo Único , Sódio , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/genética , Cálculos Urinários/genética , Urolitíase/genética
4.
Pediatr Nephrol ; 38(2): 403-415, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35552824

RESUMO

BACKGROUND: Primary hyperoxalurias (PHs) are rare genetic diseases that increase the endogenous level of oxalate, a waste metabolite excreted predominantly by the kidneys and also the gut. Treatments aim to improve oxalate excretion, or reduce oxalate generation, to prevent kidney function deterioration. Oxalobacter formigenes is an oxalate metabolizing bacterium. This Phase III, double-blind, placebo-controlled randomized trial investigated the effectiveness of orally administered Oxabact™, a lyophilized O. formigenes formulation, at reducing plasma oxalate levels in patients suffering from PH. METHODS: Subjects (≥ 2 years of age) with a diagnosis of PH and maintained but suboptimal kidney function (mean estimated glomerular filtration rate at baseline < 90 mL/min/1.73 m2) were eligible to participate. Subjects were randomized to receive Oxabact or placebo twice daily for 52 weeks. Change from baseline in plasma oxalate concentration at Week 52 was the primary study endpoint. RESULTS: Forty-three subjects were screened, 25 were recruited and one was discontinued. At Week 52, O. formigenes was established in the gut of subjects receiving Oxabact. Despite decreasing plasma oxalate level in subjects treated with Oxabact, and stable/increased levels with placebo, there was no significant difference between groups in the primary outcome (Least Squares mean estimate of treatment difference was - 3.80 µmol/L; 95% CI: - 7.83, 0.23; p-value = 0.064). Kidney function remained stable in both treatments. CONCLUSIONS: Oxabact treatment may have stabilized/reduced plasma oxalate versus a rise with placebo, but the difference over 12 months was not statistically significant (p = 0.06). A subtle effect observed with Oxabact suggests that O. formigenes may aid in preventing kidney stones. A higher resolution version of the Graphical abstract is available as Supplementary information.


Assuntos
Hiperoxalúria Primária , Hiperoxalúria , Cálculos Renais , Humanos , Hiperoxalúria/terapia , Hiperoxalúria Primária/terapia , Oxalobacter formigenes/metabolismo , Oxalatos , Cálculos Renais/metabolismo
5.
Kidney Int ; 103(1): 207-217, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36007597

RESUMO

Nedosiran is an investigational RNA interference agent designed to inhibit expression of hepatic lactate dehydrogenase, the enzyme thought responsible for the terminal step of oxalate synthesis. Oxalate overproduction is the hallmark of all genetic subtypes of primary hyperoxaluria (PH). In this double-blind, placebo-controlled study, we randomly assigned (2:1) 35 participants with PH1 (n = 29) or PH2 (n = 6) with eGFR ≥30 mL/min/1.73 m2 to subcutaneous nedosiran or placebo once monthly for 6 months. The area under the curve (AUC) of percent reduction from baseline in 24-hour urinary oxalate (Uox) excretion (primary endpoint), between day 90-180, was significantly greater with nedosiran vs placebo (least squares mean [SE], +3507 [788] vs -1664 [1190], respectively; difference, 5172; 95% CI 2929-7414; P < 0.001). A greater proportion of participants receiving nedosiran vs placebo achieved normal or near-normal (<0.60 mmol/24 hours; <1.3 × ULN) Uox excretion on ≥2 consecutive visits starting at day 90 (50% vs 0; P = 0.002); this effect was mirrored in the nedosiran-treated PH1 subgroup (64.7% vs 0; P < 0.001). The PH1 subgroup maintained a sustained Uox reduction while on nedosiran, whereas no consistent effect was seen in the PH2 subgroup. Nedosiran-treated participants with PH1 also showed a significant reduction in plasma oxalate versus placebo (P = 0.017). Nedosiran was generally safe and well tolerated. In the nedosiran arm, the incidence of injection-site reactions was 9% (all mild and self-limiting). In conclusion, participants with PH1 receiving nedosiran had clinically meaningful reductions in Uox, the mediator of kidney damage in PH.


Assuntos
Hiperoxalúria Primária , Hiperoxalúria , Humanos , Hiperoxalúria/urina , Hiperoxalúria Primária/diagnóstico , Hiperoxalúria Primária/tratamento farmacológico , Hiperoxalúria Primária/genética , Oxalatos/metabolismo , Interferência de RNA , Método Duplo-Cego
6.
Clin Kidney J ; 15(Suppl 1): i29-i32, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35711295

RESUMO

In adults, primary hyperoxaluria (PH) does not always present as obviously as in children, leading to delayed or even missed diagnosis. When diagnosed in adulthood, PH usually progresses at a slower rate and the focus is on the prevention of recurrent kidney stones as much as it is on the preservation of renal function. The most tragic presentation is when the diagnosis is made after primary non-function of a renal graft for treating previously unknown renal disease. Recurrent stones, nephrocalcinosis and features of systemic oxalosis can all be presenting features. For these reasons, consideration should be given to screening for this rare condition, using biochemical and/or genetic means, but being careful to exclude common differential diagnoses. Such efforts should be synchronized with diagnostic methods for other rare kidney diseases.

7.
Kidney Int Rep ; 7(3): 494-506, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35257062

RESUMO

Introduction: Primary hyperoxaluria type 1 (PH1) is a rare genetic disease caused by hepatic overproduction of oxalate, leading to kidney stones, nephrocalcinosis, kidney failure, and systemic oxalosis. In the 6-month double-blind period (DBP) of ILLUMINATE-A, a phase 3, randomized, placebo-controlled trial in patients with PH1 ≥6 years old, treatment with lumasiran, an RNA interference therapeutic, led to substantial reductions in urinary oxalate (UOx) levels. Methods: We report data to month 12 in the extension period (EP) of ILLUMINATE-A, including patients who continued lumasiran (lumasiran/lumasiran) or crossed over from placebo to lumasiran (placebo/lumasiran). Results: In the lumasiran/lumasiran group (n = 24), the reduction in 24-hour UOx level was sustained to month 12 (mean reduction from baseline, 66.9% at month 6; 64.1% at month 12). The placebo/lumasiran group (n = 13) had a similar time course and magnitude of 24-hour UOx reduction (mean reduction, 57.3%) after 6 months of lumasiran. Kidney stone event rates seemed to be lower after 6 months of lumasiran in both groups compared with the 12 months before consent, and this reduction was maintained at month 12 in the lumasiran/lumasiran group. At study start, 71% of patients in the lumasiran/lumasiran group and 92% in the placebo/lumasiran group had nephrocalcinosis. Nephrocalcinosis grade improved after 6 months of lumasiran in the lumasiran/lumasiran and placebo/lumasiran groups (13% and 8% of patients, respectively). After an additional 6 months of lumasiran, 46% of patients had improvement in nephrocalcinosis grade within the lumasiran/lumasiran group. Estimated glomerular filtration rate (eGFR) remained stable during the course of lumasiran treatment. The most common adverse events (AEs) related to lumasiran were mild, transient injection-site reactions (ISRs). Conclusion: Long-term lumasiran treatment enabled sustained lowering of UOx levels with acceptable safety and encouraging results on clinical outcomes.

8.
Kidney Int Rep ; 7(2): 210-220, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35155860

RESUMO

INTRODUCTION: In primary hyperoxaluria type 1 (PH1), oxalate overproduction frequently causes kidney stones, nephrocalcinosis, and kidney failure. As PH1 is caused by a congenital liver enzyme defect, combined liver-kidney transplantation (CLKT) has been recommended in patients with kidney failure. Nevertheless, systematic analyses on long-term transplantation outcomes are scarce. The merits of a sequential over combined procedure regarding kidney graft survival remain unclear as is the place of isolated kidney transplantation (KT) for patients with vitamin B6-responsive genotypes. METHODS: We used the OxalEurope registry for retrospective analyses of patients with PH1 who underwent transplantation. Analyses of crude Kaplan-Meier survival curves and adjusted relative hazards from the Cox proportional hazards model were performed. RESULTS: A total of 267 patients with PH1 underwent transplantation between 1978 and 2019. Data of 244 patients (159 CLKTs, 48 isolated KTs, 37 sequential liver-KTs [SLKTs]) were eligible for comparative analyses. Comparing CLKTs with isolated KTs, adjusted mortality was similar in patients with B6-unresponsive genotypes but lower after isolated KT in patients with B6-responsive genotypes (adjusted hazard ratio 0.07, 95% CI: 0.01-0.75, P = 0.028). CLKT yielded higher adjusted event-free survival and death-censored kidney graft survival in patients with B6-unresponsive genotypes (P = 0.025, P < 0.001) but not in patients with B6-responsive genotypes (P = 0.145, P = 0.421). Outcomes for 159 combined procedures versus 37 sequential procedures were comparable. There were 12 patients who underwent pre-emptive liver transplantation (PLT) with poor outcomes. CONCLUSION: The CLKT or SLKT remains the preferred transplantation modality in patients with PH1 with B6-unresponsive genotypes, but isolated KT could be an alternative approach in patients with B6-responsive genotypes.

9.
Kidney Int ; 100(3): 621-635, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33865885

RESUMO

Outcome data in primary hyperoxaluria type 3 (PH3), described as a less severe form of the PH's with a low risk of chronic kidney disease, are scarce. To investigate this, we retrospectively analyzed the largest PH3 cohort reported so far. Of 95 patients, 74 were followed over a median of six years. Median age of first symptoms and diagnosis were 1.9 and 6.3 years, respectively. Urolithiasis was the major clinical feature observed in 70% of pediatric and 50% of adult patients. At most recent follow-up available for 56 of the 95 patients, 21.4% were in chronic kidney disease stages 2 or more. For better characterization, samples from 49 patients were analyzed in a single laboratory and compared to data from patients with PH1 and PH2 from the same center. Urinary oxalate excretion was not significantly different from PH1 and PH2 (median: 1.37, 1.40 and 1.16 mmol/1.73m2/24hours for PH1 not responsive to vitamin B6, PH2, and PH3, respectively) but was significantly higher than in vitamin B6 responsive patients with PH1. Urinary oxalate excretion did not correlate to stone production rate nor to estimated glomerular filtration rate. Normocitraturia was present even without alkalinisation treatment; hypercalciuria was found rarely. Median plasma oxalate was significantly different only to the vitamin B6-unresponsive PH1 group. Thus, PH3 is more comparable to PH1 and PH2 than so far inferred from smaller studies. It is the most favorable PH type, but not a benign entity as it constitutes an early onset, recurrent stone disease, and kidney function can be impaired.


Assuntos
Hiperoxalúria Primária , Hiperoxalúria , Criança , Pré-Escolar , Humanos , Hiperoxalúria Primária/diagnóstico , Hiperoxalúria Primária/epidemiologia , Lactente , Oxalatos , Sistema de Registros , Estudos Retrospectivos
10.
N Engl J Med ; 384(13): 1216-1226, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33789010

RESUMO

BACKGROUND: Primary hyperoxaluria type 1 (PH1) is a rare genetic disease caused by hepatic overproduction of oxalate that leads to kidney stones, nephrocalcinosis, kidney failure, and systemic oxalosis. Lumasiran, an investigational RNA interference (RNAi) therapeutic agent, reduces hepatic oxalate production by targeting glycolate oxidase. METHODS: In this double-blind, phase 3 trial, we randomly assigned (in a 2:1 ratio) patients with PH1 who were 6 years of age or older to receive subcutaneous lumasiran or placebo for 6 months (with doses given at baseline and at months 1, 2, 3, and 6). The primary end point was the percent change in 24-hour urinary oxalate excretion from baseline to month 6 (mean percent change across months 3 through 6). Secondary end points included the percent change in the plasma oxalate level from baseline to month 6 (mean percent change across months 3 through 6) and the percentage of patients with 24-hour urinary oxalate excretion no higher than 1.5 times the upper limit of the normal range at month 6. RESULTS: A total of 39 patients underwent randomization; 26 were assigned to the lumasiran group and 13 to the placebo group. The least-squares mean difference in the change in 24-hour urinary oxalate excretion (lumasiran minus placebo) was -53.5 percentage points (P<0.001), with a reduction in the lumasiran group of 65.4% and an effect seen as early as month 1. The between-group differences for all hierarchically tested secondary end points were significant. The difference in the percent change in the plasma oxalate level (lumasiran minus placebo) was -39.5 percentage points (P<0.001). In the lumasiran group, 84% of patients had 24-hour urinary oxalate excretion no higher than 1.5 times the upper limit of the normal range at month 6, as compared with 0% in the placebo group (P<0.001). Mild, transient injection-site reactions were reported in 38% of lumasiran-treated patients. CONCLUSIONS: Lumasiran reduced urinary oxalate excretion, the cause of progressive kidney failure in PH1. The majority of patients who received lumasiran had normal or near-normal levels after 6 months of treatment. (Funded by Alnylam Pharmaceuticals; ILLUMINATE-A ClinicalTrials.gov number, NCT03681184.).


Assuntos
Hiperoxalúria Primária/tratamento farmacológico , Oxalatos/urina , RNA Interferente Pequeno/uso terapêutico , Terapêutica com RNAi , Adolescente , Adulto , Criança , Creatinina/urina , Método Duplo-Cego , Feminino , Taxa de Filtração Glomerular , Humanos , Hiperoxalúria Primária/sangue , Hiperoxalúria Primária/complicações , Hiperoxalúria Primária/urina , Cálculos Renais/prevenção & controle , Masculino , Pessoa de Meia-Idade , Oxalatos/sangue , Oxalatos/metabolismo , RNA Interferente Pequeno/efeitos adversos , Adulto Jovem
11.
Arch. esp. urol. (Ed. impr.) ; 74(1): 123-128, ene.-feb. 2021.
Artigo em Inglês | IBECS | ID: ibc-199443

RESUMO

Renal tubular acidosis (RTA) is a set of rare disorders in which the renal tubule is unable to excrete acid normally and thereby maintain normal acid-base balance, resulting in a complete or incomplete metabolic acidosis. In distal RTA (dRTA, also known as classical or type 1 RTA), there is a defect in excreting H+ ions along the distal nephron (distal tubule and collecting duct), leading to an alkaline urinary pH with calcium phosphate precipitation and stones. Causes of dRTA include genetic mutations, autoimmune disease, and some drugs. Clinical manifestations of the genetic forms of dRTA typically occur during childhood and may vary from mild clinical symptoms, such as a mild metabolic acidosis, hypokalaemia, and incidental detection of kidney stones, to more serious manifestations such as failure to thrive, severe metabolic acidosis, rickets and nephrocalcinosis. Progressive hearing loss may develop in patients with recessive dRTA, which, depending the causative gene mutation, can be present at birth or develop later in adolescence or early adulthood. Diagnosis of dRTA can be challenging, since it requires a high index of suspicion and/or measurement of urinary pH after an acid load, usually in the form of oral ammonium chloride; this should normally acidify the urine to pH below 5.3. In dRTA, urinary citrate levels are also low and patients are at increased risk of forming kidney stones from a combination of alkaline urine and low citrate. Ideally, affected patients need regular outpatient follow-up by a urologist and nephrologist. Thus, any patient found to have a calcium phosphate kidney stone, low urinary citrate, and raised urinary pH, especially with an early morning pH > 5.5, should be evaluated for underlying dRTA. Patients with complete dRTA will have a low (< 20 mmol/L) plasma or serum bicarbonate concentration, whereas in those with incomplete dRTA, bicarbonate levels are usually normal. Oral alkali as potassium citrate is still the mainstay of treatment in dRTA


La acidosis tubular renal (ATR) es un conjunto de enfermedades raras en las que el túbulo renal es incapaz de excretar ácido de forma normal y por ello de mantener un balance ácido-base normal, resultando en una acidosis metabólica completa o incompleta. En la ATR distal (ATRd, también conocida como ATR tipo 1 o clásica), hay un defecto en la excreción de iones H+ a lo largo de la parte distal de la nefrona (túbulo distal y tubo colector) que conduce a un pH urinario alcalino con precipitación de fosfato cálcico y litiasis. Las causas de la ATRd incluyen mutaciones genéticas, enfermedad autoinmune y algunos fármacos. Las manifestaciones clínicas de la forma genética de la ATRd ocurren típicamente durante la infancia y pueden variar desde síntomas leves, como acidosis metabólica leve, hipokaliemia y detección accidental de litiasis renal, hasta manifestaciones más graves tales como falta de crecimiento, acidosis metabólica severa, raquitismo y nefrocalcinosis. En pacientes con ATRd recesiva puede desarrollarse una pérdida progresiva de audición que, dependiendo de la causa de la mutación genética, puede estar presente en el momento del nacimiento o desarrollarse más tarde en la adolescencia o edad adulta temprana. El diagnóstico de la ATRd puede ser un reto ya que requiere un alto grado de sospecha y/o la medición del pH urinario tras una carga ácida, normalmente en forma de cloruro amónico oral; esto debería normalmente acidificar la orina a un pH inferior a 5,3. En la ATRd, los niveles de citrato urinario también son bajos y los pacientes tienen un mayor riesgo de formar litiasis renal por una combinación de orina alcalina e hipocitraturia. Lo ideal es que los pacientes afectados sean seguidos de forma regular por un urólogo y un nefrólogo. Así, cualquier paciente con litiasis de fosfato cálcico, hipocitraturia y pH urinario elevado, especialmente con un pH urinario matutino >5,5, debería ser estudiado para descartar una ATRd oculta. Los pacientes con ATRd completa tendrán una concentración plasmática o sérica de bicarbonato baja (< 20 mmol/L), mientras que en aquellos con una ATRd incompleta, los niveles de bicarbonato son generalmente normales. Los alcalinizantes orales como el citrato potásico son aún el principal pilar del tratamiento en la ATRd


Assuntos
Humanos , Criança , Adolescente , Adulto , Acidose Tubular Renal/diagnóstico , Acidose Tubular Renal/terapia , Cálculos Renais , Cloreto de Amônio , Ácido Cítrico , Concentração de Íons de Hidrogênio , Citrato de Potássio
12.
Arch Esp Urol ; 74(1): 123-128, 2021 Jan.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-33459628

RESUMO

Renal tubular acidosis (RTA) is a set of raredis orders in which the renal tubule is unable to excreteacid normally and there by maintain normal acid-basebalance, resulting in a complete or incomplete metabolicacidosis. In distal RTA (dRTA, also known as classicalor type 1 RTA), there is a defect in excreting H+ ionsalong the distal nephron (distal tubule and collectingduct), leading to an alkaline urinary pH with calcium phosphate precipitation and stones. Causes of dRTAinclude genetic mutations, autoimmune disease, and some drugs.Clinical manifestations of the genetic forms of dRTA typically occur during childhood and may vary from mildclinical symptoms, such as a mild metabolic acidosis, hypokalaemia,and incidental detection of kidney stones, to more serious manifestations such as failure to thrive,severe metabolic acidosis, rickets and nephrocalcinosis.Progressive hearing loss may develop in patients withrecessive dRTA, which, depending the causative genemutation, can be present at birth or develop later in adolescence or early adulthood. Diagnosis of dRTA can be challenging, since it requires a high index of suspicion and/or measurement of urinary pH after an acid load, usually in the form of oral ammonium chloride; this should normally acidify the urine to pH below 5.3. In dRTA, urinary citrate levels a real so low and patients are at increased risk of for mingkidney stones from a combination of alkaline urine and low citrate. Ideally, affected patients need regular outpatient follow-up by a urologist and nephrologist. Thus, any patient found to have a calcium phosphate kidney stone, low urinary citrate, and raised urinary pH, especially with an early morning pH >5.5, should be evaluated for underlying dRTA. Patients with complete dRTA will have a low (<20 mmol/L) plasma or serum bicarbonate concentration, whereas in those with incomplete dRTA, bicarbonate levels are usually normal. Oral alkali as potassiumcitrate is still the mainstay of treatment in dRTA.


La acidosis tubular renal (ATR) es un conjunto de enfermedades raras en las que el túbulo renal es incapaz de excretar ácido de forma normal y por ello de mantener un balance ácido-base normal, resultando en una acidosis metabólica completa o incompleta. En la ATR distal (ATRd, también conocida como ATR tipo 1 o clásica), hay un defecto en la excreción de iones H+a lo largo de la parte distal de la nefrona (túbulo distal  y tubo colector) que conduce a un pH urinario alcalino con precipitación de fosfato cálcico y litiasis. Las causas de la ATRd incluyen mutaciones genéticas, enfermedad autoinmune y algunos fármacos. Las manifestaciones clínicas de la forma genética de la ATRd ocurren típicamente durante la infancia y pueden variar desde síntomas leves, como acidosis metabólica leve, hipokaliemia y detección accidental de litiasis renal, hasta manifestaciones más graves tales como falta de crecimiento, acidosis metabólica severa, raquitismo y nefrocalcinosis. En pacientes con ATRd recesiva puede desarrollarse una pérdida progresiva de audición que, dependiendo de la causa de la mutación genética, puede estar presente en el momento del nacimientoo desarrollarse más tarde en la adolescencia o edad adulta temprana.El diagnóstico de la ATRd puede ser un reto ya que requiere un alto grado de sospecha y/o la medición del pH urinario tras una carga ácida, normalmente en forma de cloruro amónico oral; esto debería normalmente acidificar la orina a un pH inferior a 5,3. En la ATRd, los niveles de citrato urinario también son bajos y los pacientes tienen un mayor riesgo de formar litiasis renal por una combinación de orina alcalina e hipocitraturia. Lo ideal es que los pacientes afectados sean seguidos de forma regular por un urólogo y un nefrólogo. Así,cualquier paciente con litiasis de fosfato cálcico, hipocitraturia y pH urinario elevado, especialmente con un pH urinario matutino >5,5, debería ser estudiado para descartar una ATRd oculta. Los pacientes con ATRd completa tendrán una concentración plasmática o sérica de bicarbonato baja (<20 mmol/L), mientras que en aquellos con una ATRd incompleta, los niveles de bicarbonato son generalmente normales. Los alcalinizantes orales como el citrato potásico son aún el principal pilar del tratamiento en la ATRd.


Assuntos
Acidose Tubular Renal , Cálculos Renais , Acidose Tubular Renal/diagnóstico , Acidose Tubular Renal/terapia , Adolescente , Adulto , Cloreto de Amônio , Criança , Ácido Cítrico , Humanos , Concentração de Íons de Hidrogênio
13.
Pediatr Nephrol ; 32(5): 781-790, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27924398

RESUMO

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.


Assuntos
Hiperoxalúria/terapia , Oxalobacter formigenes , Adolescente , Carga Bacteriana , Criança , Pré-Escolar , Método Duplo-Cego , Fezes/microbiologia , Feminino , Humanos , Hiperoxalúria/fisiopatologia , Hiperoxalúria/urina , Testes de Função Renal , Masculino , Ácido Oxálico/urina , Probióticos/administração & dosagem , Probióticos/efeitos adversos , Probióticos/uso terapêutico , Comprimidos com Revestimento Entérico , Resultado do Tratamento , Adulto Jovem
14.
Sci Rep ; 6: 34737, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27721432

RESUMO

Cystinuria is the commonest inherited cause of nephrolithiasis (~1% in adults; ~6% in children) and is the result of impaired cystine reabsorption in the renal proximal tubule. Cystine is poorly soluble in urine with a solubility of ~1 mM and can readily form microcrystals that lead to cystine stone formation, especially at low urine pH. Diagnosis of cystinuria is made typically by ion-exchange chromatography (IEC) detection and quantitation, which is slow, laboursome and costly. More rapid and frequent monitoring of urinary cystine concentration would significantly improve the diagnosis and clinical management of cystinuria. We used attenuated total reflection - Fourier transform infrared spectroscopy (ATR-FTIR) to detect and quantitate insoluble cystine in 22 cystinuric and 5 healthy control urine samples. Creatinine concentration was also determined by ATR-FTIR to adjust for urinary concentration/dilution. Urine was centrifuged, the insoluble fraction re-suspended in 5 µL water and dried on the ATR prism. Cystine was quantitated using its 1296 cm-1 absorption band and levels matched with parallel measurements made using IEC. ATR-FTIR afforded a rapid and inexpensive method of detecting and quantitating insoluble urinary cystine. This proof-of-concept study provides a basis for developing a high-throughput, cost-effective diagnostic method for cystinuria, and for point-of-care clinical monitoring.


Assuntos
Cistinúria/diagnóstico , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Urina/química , Creatinina/urina , Cistinúria/urina , Ensaios de Triagem em Larga Escala , Humanos , Sistemas Automatizados de Assistência Junto ao Leito , Sensibilidade e Especificidade , Espectroscopia de Infravermelho com Transformada de Fourier/economia
15.
Clin J Am Soc Nephrol ; 10(2): 278-85, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25635036

RESUMO

BACKGROUND AND OBJECTIVES: Recent epidemiologic studies have provided evidence for an association between nephrolithiasis and cardiovascular disease, although the underlying mechanism is still unclear. Vascular calcification (VC) is a strong predictor of cardiovascular morbidity and the hypothesis explored in this study is that VC is more prevalent in calcium kidney stone formers (KSFs). The aims of this study were to determine (1) whether recurrent calcium KSFs have more VC and osteoporosis compared with controls and (2) whether hypercalciuria is related to VC in KSFs. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: This is a retrospective, matched case-control study that included KSFs attending an outpatient nephrology clinic of the Royal Free Hospital (London, UK) from 2011 to 2014. Age- and sex-matched non-stone formers were drawn from a list of potential living kidney donors from the same hospital. A total of 111 patients were investigated, of which 57 were KSFs and 54 were healthy controls. Abdominal aortic calcification (AAC) and vertebral bone mineral density (BMD) were assessed using available computed tomography (CT) imaging. The prevalence, severity, and associations of AAC and CT BMD between KSFs and non-stone formers were compared. RESULTS: Mean age was 47±14 years in KSFs and 47±13 in non-stone formers. Men represented 56% and 57% of KSFs and non-stone formers, respectively. The prevalence of AAC was similar in both groups (38% in KSFs versus 35% in controls, P=0.69). However, the AAC severity score (median [25th percentile, 75th percentile]) was significantly higher in KSFs compared with the control group (0 [0, 43] versus 0 [0, 10], P<0.001). In addition, the average CT BMD was significantly lower in KSFs (159±53 versus 194 ±48 Hounsfield units, P<0.001). A multivariate model adjusted for age, sex, high BP, diabetes, smoking status, and eGFR confirmed that KSFs have higher AAC scores and lower CT BMD compared with non-stone formers (P<0.001 for both). Among stone formers, the association between AAC score and hypercalciuria was not statistically significant (P=0.86). CONCLUSIONS: This study demonstrates that patients with calcium kidney stones suffer from significantly higher degrees of aortic calcification than age- and sex-matched non-stone formers, suggesting that VC may be an underlying mechanism explaining reported associations between nephrolithiasis and cardiovascular disease. Moreover, bone demineralization is more prominent in KSFs. However, more data are needed to confirm the possibility of potentially common underlying mechanisms leading to extraosseous calcium deposition and osteoporosis in KSFs.


Assuntos
Doenças da Aorta/epidemiologia , Densidade Óssea , Cálculos Renais/epidemiologia , Osteoporose/epidemiologia , Calcificação Vascular/epidemiologia , Adulto , Aorta Abdominal/diagnóstico por imagem , Doenças da Aorta/diagnóstico , Aortografia/métodos , Distribuição de Qui-Quadrado , Feminino , Humanos , Hipercalciúria/diagnóstico , Hipercalciúria/epidemiologia , Cálculos Renais/diagnóstico , Modelos Lineares , Londres/epidemiologia , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Osteoporose/diagnóstico , Ambulatório Hospitalar , Prevalência , Recidiva , Estudos Retrospectivos , Fatores de Risco , Índice de Gravidade de Doença , Coluna Vertebral/diagnóstico por imagem , Tomografia Computadorizada por Raios X , Calcificação Vascular/diagnóstico
16.
Physiol Rep ; 1(6): e00160, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24400161

RESUMO

A multiplex family was identified with biochemical and clinical features suggestive of Bartter's syndrome (BS). The eldest sibling presented with developmental delay and rickets at 4 years of age with evidence of hypercalciuria and hypokalemia. The second sibling presented at 1 year of age with urinary tract infections, polyuria, and polydipsia. The third child was born after a premature delivery with a history of polyhydramnios and neonatal hypocalcemia. Following corrective treatment she also developed hypercalciuria and a hypokalemic metabolic alkalosis. There was evidence of secondary hyperreninemia and hyperaldosteronism in all three siblings consistent with BS. Known BS genes were screened and functional assays of ROMK (alias KCNJ1, Kir1.1) were carried out in Xenopus oocytes. We detected compound heterozygous missense changes in KCNJ1, encoding the potassium channel ROMK. The S219R/L220F mutation was segregated from father and mother, respectively. In silico modeling of the missense mutations suggested deleterious changes. Studies in Xenopus oocytes revealed that both S219R and L220F had a deleterious effect on ROMK-mediated potassium currents. Coinjection to mimic the compound heterozygosity produced a synergistic decrease in channel function and revealed a loss of PKA-dependent stabilization of PIP2 binding. In conclusion, in a multiplex family with BS, we identified compound heterozygous mutations in KCNJ1. Functional studies of ROMK confirmed the pathogenicity of these mutations and defined the mechanism of channel dysfunction.

17.
Nephron Physiol ; 120(4): p33-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23075758

RESUMO

Pyrophosphate (PPi) is well known as a regulator of calcification, and the ANKH (ANK in mouse) protein has a role in the membrane transport of PPi. Earlier work concentrated on bones and joints, but ANKH is also likely to have important roles in the kidney, with newer studies focusing on vascular calcification in renal failure. Renal calcification can occur due to a naturally occurring ANK mouse mutation, yet other ANK mutations do not cause a renal phenotype. Despite evidence over 10 years of ANKH's involvement in PPi transport, efflux of PPi via ANKH has never been demonstrated. Rather than physically moving PPi, the ANKH protein may assist its membrane transport in other ways such as by hydrolysis and compartmentalisation. Protein complexes may account for effects of ANKH that are specific to particular tissues. In the kidney, recent localisation data may be helpful in suggesting physiological roles for ANKH, such as its co-localisation with aquaporin-2 and cilial proteins. Such diverse functions would reflect the ubiquitous nature of ANKH in tissues and its profound evolutionary conservation.


Assuntos
Difosfatos/metabolismo , Rim/metabolismo , Proteínas de Transporte de Fosfato/genética , Proteínas de Transporte de Fosfato/metabolismo , Animais , Aquaporina 2/genética , Aquaporina 2/metabolismo , Calcinose/metabolismo , Cílios/metabolismo , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Mutação , Nefrocalcinose/metabolismo , Especificidade de Órgãos , Insuficiência Renal/metabolismo
18.
Cell Physiol Biochem ; 24(5-6): 595-604, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19910700

RESUMO

BACKGROUND/AIMS: ANKH encodes a putative pyrophosphate transporter named ANKH, which regulates tissue calcification. ANKH is a transmembrane protein with at least 8 predicted transmembrane domains. Sequence analysis reveals a possible cilial localisation motif immediately after the last transmembrane segment. Here we aim to determine the subcellular localisation of ANKH in ciliated epithelial cells and murine tissue and identify colocalisation using ciliary/basal body markers. METHODS: Using murine kidney, renal epithelial cells and osteoblast cells we investigated the expression and localisation of ANKH using RT-PCR, Western blotting and immunocytochemistry. RESULTS: Here we confirm endogenous expression of ANKH mRNA and protein in whole mouse kidney as well as mouse renal epithelial cell lines M1 and mpkCCDcl4 and the osteoblast cell line MC3T3-E1. Using antibodies directed towards ANKH, we confirm cilial and basal body localisation in renal tissues and renal epithelial cells, in addition to a centrosomal localisation in dividing mpkCCDcl4 cells. We also establish that the osteoblast cell line MC3T3-E1 forms an epithelioid cell layer, with junctional complex formation and primary cilia expression. ANKH is also seen within cilial and basal body structures of MC3T3-E1 cells. An ANKH-3XFLAG construct expressed in mpkCCDcl4 cells also localises to the primary cilium/basal body complex confirming this localisation. CONCLUSION: We conclude that the transmembrane protein ANKH is expressed in cilia and basal body structures, and postulate a sensory role at this location.


Assuntos
Proteínas de Transporte de Ânions/análise , Osso e Ossos/metabolismo , Cílios/metabolismo , Rim/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Linhagem Celular , Humanos , Camundongos , Dados de Sequência Molecular , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
19.
Biochem Biophys Res Commun ; 371(4): 877-82, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18477472

RESUMO

Nephronophthisis is characterised by renal fibrosis, tubular basement membrane disruption and corticomedullary cyst formation leading to end stage renal failure. Mutations in NPHP1 account for the underlying genetic defect in 25% of patients with nephronophthisis. Loss of urine concentration ability may be an early feature of nephronophthisis. Using yeast-2-library screening with the SH3 domain of nephrocystin-1 as bait, we identify Ack1 as a novel interaction partner. This interaction is confirmed using exogenous over-expression followed by co-immunoprecipitation. Ack1 is an activated Cdc42-associated kinase, and like nephrocystin-1, is a known interactor of p130Cas. Nephrocystin-1 partially colocalises with Ack1 at cell-cell contacts in IMCD3 cells. In human kidney, nephrocystin-1 expression is limited to cell-cell junctions in renal collecting duct cells. These data define Ack1 as a novel interaction partner of nephrocystin-1 and implicate cell-cell junctions and the renal collecting duct in the pathology of nephronophthisis.


Assuntos
Túbulos Renais Coletores/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Linhagem Celular , Proteínas do Citoesqueleto , Humanos , Imunoprecipitação , Junções Intercelulares/metabolismo , Túbulos Renais Coletores/citologia , Proteínas de Membrana , Proteínas/genética , Técnicas do Sistema de Duplo-Híbrido , Domínios de Homologia de src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...