Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Urolithiasis ; 50(2): 119-130, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35122487

RESUMO

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


Assuntos
Hiperoxalúria Primária , Hiperoxalúria , Nefrocalcinose , Animais , Oxalato de Cálcio , Humanos , Hiperoxalúria/complicações , Hiperoxalúria Primária/complicações , Nefrocalcinose/complicações , Nefrocalcinose/prevenção & controle , Oxalatos/urina , Oxalobacter formigenes , Ratos
2.
Surg Today ; 52(1): 12-21, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33464414

RESUMO

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


Assuntos
Síndrome do Intestino Curto/reabilitação , Síndrome do Intestino Curto/terapia , Fatores Etários , Doenças Ósseas Metabólicas/etiologia , Doenças Ósseas Metabólicas/prevenção & controle , Pré-Escolar , Feminino , Peptídeo 2 Semelhante ao Glucagon/uso terapêutico , Humanos , Incidência , Lactente , Recém-Nascido , Insuficiência Intestinal/etiologia , Insuficiência Intestinal/prevenção & controle , Intestino Delgado/transplante , Hepatopatias/etiologia , Hepatopatias/prevenção & controle , Masculino , Nefrocalcinose/etiologia , Nefrocalcinose/prevenção & controle , Qualidade de Vida , Síndrome do Intestino Curto/epidemiologia , Síndrome do Intestino Curto/etiologia
3.
Endocrinol Diabetes Metab ; 4(2): e00185, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33855198

RESUMO

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


Assuntos
Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/cirurgia , Eletrólitos/metabolismo , Transplante de Rim , Inibidores do Transportador 2 de Sódio-Glicose/administração & dosagem , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/prevenção & controle , Adulto , Idoso , Feminino , Seguimentos , Humanos , Hipercalciúria/etiologia , Hipercalciúria/prevenção & controle , Transplante de Rim/efeitos adversos , Masculino , Pessoa de Meia-Idade , Nefrocalcinose/etiologia , Nefrocalcinose/prevenção & controle , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/prevenção & controle , Erros Inatos do Transporte Tubular Renal/etiologia , Erros Inatos do Transporte Tubular Renal/prevenção & controle , Estudos Retrospectivos , Risco , Fatores de Tempo , Resultado do Tratamento , Aumento de Peso
4.
J Pediatr Endocrinol Metab ; 34(6): 791-798, 2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-33837680

RESUMO

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


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Substituição de Medicamentos/métodos , Raquitismo Hipofosfatêmico Familiar/tratamento farmacológico , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Hidroxicolecalciferóis/uso terapêutico , Nefrocalcinose/prevenção & controle , Conservadores da Densidade Óssea/uso terapêutico , Criança , Raquitismo Hipofosfatêmico Familiar/patologia , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/imunologia , Seguimentos , Humanos , Injeções Intravenosas , Masculino , Prognóstico , Estudos Retrospectivos
5.
Cancer Chemother Pharmacol ; 86(3): 383-391, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32789758

RESUMO

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


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Hipercalciúria/prevenção & controle , Magnésio/administração & dosagem , Recidiva Local de Neoplasia/tratamento farmacológico , Nefrocalcinose/prevenção & controle , Erros Inatos do Transporte Tubular Renal/prevenção & controle , Adulto , Idoso , Idoso de 80 Anos ou mais , Cetuximab/administração & dosagem , Neoplasias Colorretais/patologia , Feminino , Seguimentos , Humanos , Hipercalciúria/induzido quimicamente , Hipercalciúria/patologia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Nefrocalcinose/induzido quimicamente , Nefrocalcinose/patologia , Panitumumabe/administração & dosagem , Prognóstico , Erros Inatos do Transporte Tubular Renal/induzido quimicamente , Erros Inatos do Transporte Tubular Renal/patologia , Estudos Retrospectivos
7.
Exp Cell Res ; 376(2): 210-220, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30690028

RESUMO

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


Assuntos
Antineoplásicos Imunológicos/efeitos adversos , Carcinoma de Células Escamosas/tratamento farmacológico , Cetuximab/efeitos adversos , Inibidores do Crescimento/uso terapêutico , Imidazóis/uso terapêutico , Neoplasias Bucais/tratamento farmacológico , Piridinas/uso terapêutico , Animais , Carcinoma de Células Escamosas/complicações , Linhagem Celular Tumoral , Quimioterapia Combinada , Receptores ErbB/antagonistas & inibidores , Exantema/induzido quimicamente , Exantema/genética , Exantema/prevenção & controle , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Inibidores do Crescimento/efeitos adversos , Inibidores do Crescimento/antagonistas & inibidores , Células HEK293 , Humanos , Hipercalciúria/induzido quimicamente , Hipercalciúria/genética , Hipercalciúria/prevenção & controle , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Bucais/complicações , Neoplasias Bucais/genética , Nefrocalcinose/induzido quimicamente , Nefrocalcinose/genética , Nefrocalcinose/prevenção & controle , Erros Inatos do Transporte Tubular Renal/induzido quimicamente , Erros Inatos do Transporte Tubular Renal/genética , Erros Inatos do Transporte Tubular Renal/prevenção & controle , Transcriptoma , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Urolithiasis ; 47(1): 79-89, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30535828

RESUMO

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


Assuntos
Testes Genéticos/métodos , Hiperoxalúria Primária/genética , Cálculos Renais/prevenção & controle , Nefrocalcinose/prevenção & controle , Oxalatos/metabolismo , Oxirredutases do Álcool/genética , Hidratação , Humanos , Hiperoxalúria Primária/complicações , Hiperoxalúria Primária/diagnóstico , Hiperoxalúria Primária/terapia , Cálculos Renais/etiologia , Cálculos Renais/cirurgia , Transplante de Rim , Litotripsia , Transplante de Fígado , Mutação , Nefrocalcinose/etiologia , Oxo-Ácido-Liases/genética , Piridoxina/uso terapêutico , Terapia de Substituição Renal/métodos , Transaminases/genética , Resultado do Tratamento
9.
Eur J Med Genet ; 62(11): 103577, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30423445

RESUMO

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


Assuntos
Hipercalcemia/diagnóstico , Hipercalcemia/genética , Doenças do Recém-Nascido/diagnóstico , Doenças do Recém-Nascido/genética , Erros Inatos do Metabolismo/diagnóstico , Erros Inatos do Metabolismo/genética , Patologia Molecular , Vitamina D3 24-Hidroxilase/genética , Adolescente , Criança , Pré-Escolar , Variações do Número de Cópias de DNA/genética , Feminino , Humanos , Hipercalcemia/tratamento farmacológico , Hipercalcemia/patologia , Lactente , Doenças do Recém-Nascido/tratamento farmacológico , Doenças do Recém-Nascido/patologia , Mutação com Perda de Função/genética , Masculino , Erros Inatos do Metabolismo/tratamento farmacológico , Erros Inatos do Metabolismo/patologia , Nefrocalcinose/tratamento farmacológico , Nefrocalcinose/genética , Nefrocalcinose/prevenção & controle , Insuficiência Renal/tratamento farmacológico , Insuficiência Renal/genética , Insuficiência Renal/prevenção & controle , Deleção de Sequência/genética , Vitamina D/uso terapêutico
10.
Nat Commun ; 9(1): 5454, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30575740

RESUMO

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


Assuntos
Oxirredutases do Álcool/antagonistas & inibidores , Sistemas CRISPR-Cas , Terapia Genética/métodos , Hiperoxalúria Primária/terapia , Oxalatos/urina , Oxirredutases do Álcool/genética , Animais , Modelos Animais de Doenças , Edição de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Nefrocalcinose/prevenção & controle
11.
J Trace Elem Med Biol ; 50: 327-331, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30262299

RESUMO

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


Assuntos
Quimiorradioterapia/efeitos adversos , Hipercalciúria/prevenção & controle , Hipocalcemia/prevenção & controle , Sulfato de Magnésio/uso terapêutico , Nefrocalcinose/prevenção & controle , Erros Inatos do Transporte Tubular Renal/prevenção & controle , Cetuximab/efeitos adversos , Cetuximab/uso terapêutico , Cisplatino/efeitos adversos , Cisplatino/uso terapêutico , Feminino , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Humanos , Masculino , Estudos Prospectivos
12.
J Am Soc Nephrol ; 29(6): 1636-1648, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29654213

RESUMO

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


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Falência Renal Crônica/sangue , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Calcificação Vascular/prevenção & controle , Sulfato de Zinco/farmacologia , Animais , Aorta , Transdiferenciação Celular , Células Cultivadas , Suplementos Nutricionais , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Glucuronidase/genética , Humanos , Hidroxietilrutosídeo , Hiperfosfatemia/sangue , Hiperfosfatemia/complicações , Proteínas Klotho , Camundongos , NF-kappa B/antagonistas & inibidores , Nefrectomia , Nefrocalcinose/prevenção & controle , Fosfatos , Transdução de Sinais , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Calcificação Vascular/sangue , Calcificação Vascular/etiologia , Zinco/sangue
13.
Kidney Int ; 93(3): 656-669, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29241624

RESUMO

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.


Assuntos
Plasticidade Celular , Hiperoxalúria/metabolismo , Inflamassomos/metabolismo , Interleucina-1/metabolismo , Rim/metabolismo , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Nefrocalcinose/metabolismo , Insuficiência Renal Crônica/metabolismo , Animais , Butileno Glicóis/farmacologia , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Plasticidade Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Hiperoxalúria/tratamento farmacológico , Hiperoxalúria/imunologia , Hiperoxalúria/patologia , Inflamassomos/efeitos dos fármacos , Inflamassomos/genética , Inflamassomos/imunologia , Interleucina-1/imunologia , Rim/imunologia , Rim/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Nefrocalcinose/imunologia , Nefrocalcinose/patologia , Nefrocalcinose/prevenção & controle , Fenótipo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Insuficiência Renal Crônica/imunologia , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/prevenção & controle , Transdução de Sinais
14.
Kidney Int ; 93(3): 580-588, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29129401

RESUMO

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


Assuntos
Claudinas/deficiência , Hipercalciúria/prevenção & controle , Túbulos Renais Distais/metabolismo , Alça do Néfron/metabolismo , Deficiência de Magnésio/prevenção & controle , Animais , Cálcio/metabolismo , Claudinas/genética , Modelos Animais de Doenças , Deleção de Genes , Predisposição Genética para Doença , Hipercalciúria/genética , Hipercalciúria/metabolismo , Hipercalciúria/fisiopatologia , Túbulos Renais Distais/patologia , Túbulos Renais Distais/fisiopatologia , Alça do Néfron/patologia , Alça do Néfron/fisiopatologia , Magnésio/metabolismo , Deficiência de Magnésio/genética , Deficiência de Magnésio/metabolismo , Deficiência de Magnésio/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrocalcinose/genética , Nefrocalcinose/metabolismo , Nefrocalcinose/fisiopatologia , Nefrocalcinose/prevenção & controle , Fenótipo , Sódio/metabolismo
15.
Trials ; 18(1): 424, 2017 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-28893306

RESUMO

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


Assuntos
Colecalciferol/administração & dosagem , Suplementos Nutricionais , Recém-Nascido Prematuro , Nascimento Prematuro , Deficiência de Vitamina D/tratamento farmacológico , Biomarcadores/sangue , Doenças Ósseas Metabólicas/epidemiologia , Doenças Ósseas Metabólicas/prevenção & controle , Colecalciferol/efeitos adversos , Protocolos Clínicos , Suplementos Nutricionais/efeitos adversos , Idade Gestacional , Humanos , Incidência , Lactente , Recém-Nascido , Recém-Nascido Prematuro/sangue , Nefrocalcinose/epidemiologia , Nefrocalcinose/prevenção & controle , Nefrolitíase/epidemiologia , Nefrolitíase/prevenção & controle , Polônia/epidemiologia , Nascimento Prematuro/sangue , Projetos de Pesquisa , Fatores de Tempo , Resultado do Tratamento , Vitamina D/análogos & derivados , Vitamina D/sangue , Deficiência de Vitamina D/sangue , Deficiência de Vitamina D/diagnóstico , Deficiência de Vitamina D/epidemiologia
16.
Pediatr Diabetes ; 18(5): 327-331, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28568353

RESUMO

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


Assuntos
Doença de Depósito de Glicogênio Tipo I/terapia , Medicina de Precisão , Insuficiência Renal/prevenção & controle , Adenoma/complicações , Adenoma/prevenção & controle , Adulto , Criança , Terapia Combinada , Doença de Depósito de Glicogênio Tipo I/sangue , Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/diagnóstico , Humanos , Hipoglicemia/complicações , Hipoglicemia/prevenção & controle , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/prevenção & controle , Nefrocalcinose/complicações , Nefrocalcinose/prevenção & controle , Nefrolitíase/complicações , Nefrolitíase/prevenção & controle , Osteoporose/complicações , Osteoporose/prevenção & controle , Prognóstico , Insuficiência Renal/complicações
17.
Yakugaku Zasshi ; 137(1): 79-82, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-28049899

RESUMO

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


Assuntos
Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Hipercalciúria/induzido quimicamente , Hipercalciúria/epidemiologia , Nefrocalcinose/induzido quimicamente , Nefrocalcinose/epidemiologia , Erros Inatos do Transporte Tubular Renal/induzido quimicamente , Erros Inatos do Transporte Tubular Renal/epidemiologia , Neoplasias do Colo do Útero/tratamento farmacológico , Antineoplásicos/toxicidade , Cisplatino/toxicidade , Terapia Combinada , Feminino , Humanos , Hipercalciúria/prevenção & controle , Incidência , Monitorização Fisiológica , Nefrocalcinose/prevenção & controle , Erros Inatos do Transporte Tubular Renal/prevenção & controle , Estudos Retrospectivos , Neoplasias do Colo do Útero/radioterapia
18.
Endocr J ; 64(3): 283-289, 2017 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-28025445

RESUMO

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


Assuntos
Raquitismo Hipofosfatêmico Familiar/fisiopatologia , Hiperparatireoidismo Secundário/etiologia , Hipertensão/etiologia , Nefrocalcinose/etiologia , Adolescente , Adulto , Idade de Início , Conservadores da Densidade Óssea/uso terapêutico , Criança , Pré-Escolar , Suplementos Nutricionais , Raquitismo Hipofosfatêmico Familiar/dietoterapia , Raquitismo Hipofosfatêmico Familiar/genética , Feminino , Hospitais Pediátricos , Humanos , Hidroxicolecalciferóis/uso terapêutico , Hiperparatireoidismo Secundário/epidemiologia , Hiperparatireoidismo Secundário/prevenção & controle , Hipertensão/epidemiologia , Hipertensão/prevenção & controle , Masculino , Prontuários Médicos , Mutação , Nefrocalcinose/epidemiologia , Nefrocalcinose/prevenção & controle , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Fosfatos/uso terapêutico , Prevalência , Estudos Retrospectivos , Tóquio/epidemiologia , Adulto Jovem
19.
Kidney Int ; 89(5): 1027-1036, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27083280

RESUMO

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


Assuntos
Aorta Abdominal/metabolismo , Aorta Torácica/metabolismo , Doenças da Aorta/prevenção & controle , Rim/metabolismo , Nefrocalcinose/prevenção & controle , Osteopontina/metabolismo , Fosfatos , Calcificação Vascular/prevenção & controle , Animais , Aorta Abdominal/patologia , Aorta Torácica/patologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Biomarcadores/sangue , Remodelação Óssea , Dieta , Modelos Animais de Doenças , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Rim/patologia , Camundongos Endogâmicos DBA , Camundongos Knockout , Nefrocalcinose/genética , Nefrocalcinose/metabolismo , Nefrocalcinose/patologia , Osteopontina/deficiência , Osteopontina/genética , Calcificação Vascular/genética , Calcificação Vascular/metabolismo , Calcificação Vascular/patologia
20.
Internist (Berl) ; 55(2): 199-205, 2014 Feb.
Artigo em Alemão | MEDLINE | ID: mdl-24419536

RESUMO

We report about a female patient with severe hypomagnesemia under therapy with proton pump inhibitors (PPI) who presented with a cerebral seizure. Chronic use of PPIs can cause hypomagnesemia. Because of mostly unspecific symptoms which become symptomatic only with severe deficiency, the disease pattern is underdiagnosed. Hypomagnesemia is currently coming increasingly more to the forefront of medical literature.


Assuntos
Apraxias/induzido quimicamente , Tontura/induzido quimicamente , Hipercalciúria/induzido quimicamente , Hipercalciúria/diagnóstico , Nefrocalcinose/induzido quimicamente , Nefrocalcinose/diagnóstico , Inibidores da Bomba de Prótons/efeitos adversos , Erros Inatos do Transporte Tubular Renal/induzido quimicamente , Erros Inatos do Transporte Tubular Renal/diagnóstico , Convulsões/induzido quimicamente , Idoso , Apraxias/prevenção & controle , Diagnóstico Diferencial , Tontura/prevenção & controle , Feminino , Humanos , Hipercalciúria/prevenção & controle , Nefrocalcinose/prevenção & controle , Erros Inatos do Transporte Tubular Renal/prevenção & controle , Convulsões/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA