Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Cell Signal ; 112: 110887, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37717713

RESUMO

Sirtuin1 (Sirt1) activation significantly attenuated calcium oxalate (CaOx) crystal deposition and renal inflammatory injury by regulating renal immune microenvironment. Here, to elucidate the molecular mechanism underlying the therapeutic effects of Sirt1 on macrophage related inflammation and tubular epithelial cells (TECs) necrosis, we constructed a macrophage and CaOx monohydrate (COM)-stimulated tubular cell co-culture system to mimic immune microenvironment in kidney and established a mouse model of CaOx nephrocalcinosis in wild-type and myeloid-specific Sirt1 knockout mice. Target prediction analyses of Gene Expression Omnibus Datasets showed that only miR-34b-5p is regulated by lipopolysaccharides and upregulated by SRT1720 and targets the TLR4 3'-untranslated region. In vitro, SRT1720 suppressed TLR4 expression and M1 macrophage polarization and decreased reactive oxygen species (ROS) production and mitochondrial damage in COM-stimulated TECs by targeting miR-34b-5p. Mechanically, Sirt1 promoted miR-34b-5p expression by suppressing the tri-methylation of H3K27, which directly bound to the miR-34b-5p promoter and abolished the miR-34b-5p transcription. Furthermore, loss of Sirt1 aggravated CaOx nephrocalcinosis-induced inflammatory and oxidative kidney injury, while AgomiR-34b reversed these effects. Therefore, our data suggested that Sirt1 inhibited TLR4 signaling and M1 macrophage polarization and decreased inflammatory and oxidative injury of TECs in vitro and in vivo.


Assuntos
MicroRNAs , Nefrocalcinose , Camundongos , Animais , Oxalato de Cálcio/metabolismo , Oxalato de Cálcio/farmacologia , Nefrocalcinose/metabolismo , Sirtuína 1/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Rim/metabolismo , Macrófagos/metabolismo
2.
Antioxid Redox Signal ; 38(10-12): 731-746, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36242511

RESUMO

Aims: Calcium oxalate (CaOx) crystal deposition induces damage to the renal tubular epithelium, increases epithelial adhesion, and contributes to CaOx nephrocalcinosis. The long noncoding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) is thought to be involved in this process. In this study, we aimed to investigate the mechanism by which NEAT1 regulates renal tubular epithelium in response to inflammatory and oxidative injury triggered by CaOx crystals. Results: As CaOx crystals were deposited in mouse kidney tissue, the expression of NEAT1 was significantly elevated and positively correlated with interferon regulatory factor 1 (IRF1), Toll-like receptor 4 (TLR4), and NF-κB. NEAT1 targets and inhibits miR-130a-3p as a competitor to endogenous RNA. miR-130 binds to and exerts inhibitory effects on the 3'-untranslated region of IRF1. After transfected with silence-NEAT1, IRF1, TLR4, and NF-κB were also variously inhibited, and oxidative damage in renal calcinosis was subsequently attenuated. When we simultaneously inhibited NEAT1 and miR-130, renal tubular injury was exacerbated. Innovation and Conclusion: We found that the lncRNA NEAT1 can enhance IRF1 signaling through targeted repression of miR-130a-3p and activate TLR4/NF-κB pathways to promote oxidative damage during CaOx crystal deposition. This provides an explanation for the tubular epithelial damage caused by CaOx crystals and offers new ideas and drug targets for the prevention and treatment of CaOx nephrocalcinosis. Antioxid. Redox Signal. 38, 731-746.


Assuntos
Calcinose , MicroRNAs , Nefrocalcinose , RNA Longo não Codificante , Camundongos , Animais , Oxalato de Cálcio/química , Oxalato de Cálcio/metabolismo , Oxalato de Cálcio/farmacologia , Nefrocalcinose/metabolismo , Receptor 4 Toll-Like/metabolismo , RNA Longo não Codificante/genética , NF-kappa B/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Fator Regulador 1 de Interferon/farmacologia , Rim/metabolismo , Estresse Oxidativo , MicroRNAs/genética , Oxirredução
3.
Oxid Med Cell Longev ; 2021: 1676152, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34512861

RESUMO

The roles of the lncRNA X inactive specific transcript (XIST) in many diseases, including cancers and inflammatory sickness, have been previously elucidated. However, renal calculus remained poorly understood. In this study, we revealed the potential effects of XIST on kidney stones that were exerted via inflammatory response and oxidative stress mechanisms. We established a glyoxylate-induced calcium oxalate (CaOx) stone mouse model and exposed HK-2 cells to calcium oxalate monohydrate (COM). The interactions among XIST, miR-223-3p, and NOD-like receptor protein 3 (NLRP3) and their respective effects were determined by RNAs and protein expression, luciferase activity, and immunohistochemistry (IHC) assays. Cell necrosis, reactive oxygen species (ROS) generation, and inflammatory responses were detected after silencing XIST, activating and inhibiting miR-223-3p, and both knocking down XIST and activating miR-223-3p in vitro and in vivo. The XIST, NLRP3, caspase-1, and IL-1ß levels were notably increased in kidney samples from glyoxylate-induced CaOx stone model mice. XIST knockdown significantly suppressed the inflammatory damage and ROS production and further attenuated oxalate crystal deposition. miRNA-223-3p mimics also exerted the same effects. Moreover, we verified the interactions among XIST, miRNA-223-3p and NLRP3, and the subsequent effects. Our results suggest that the lncRNA XIST participates in the formation and progression of renal calculus by interacting with miR-223-3p and the NLRP3/Caspase-1/IL-1ß pathway to mediate the inflammatory response and ROS production.


Assuntos
MicroRNAs/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Nefrocalcinose/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , RNA Longo não Codificante/antagonistas & inibidores , Animais , Oxalato de Cálcio/administração & dosagem , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nefrocalcinose/metabolismo , Nefrocalcinose/patologia , Transfecção
4.
Regul Toxicol Pharmacol ; 117: 104776, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32871170

RESUMO

When extrapolating data from animal toxicological studies a default factor (dUF) of 100 is applied to derive a heath based guidance value. The UF takes into account the interspecies differences (ID) and the intraspecies variability (IV). When re-evaluating the safety of phosphates used as food additives nephrocalcinosis was identified as the critical endpoint. The underlying mechanism for nephrocalcinosis was attributed to the precipitation of calcium phosphate in the kidney, depending on its solubility, irrespective of the species and the population. Based on the mechanism, the volume of primary urine, for which the glomerular filtration rate (GFR) was used as a proxy, was considered to be the only parameter relevant for ID and IV. Median value of GFR in rats was 4.0 ml/min/kg bw. In humans it was 1.6 ml/min/kg bw in healthy adults and 0.9 in elderly. These values were calculated from the distribution of the GFR data from 8 studies in rats (n = 191), 16 studies in adults (n = 1540) and 5 studies in elderly (n = 2608). Multiplying the distribution of the ratio rat/healthy humans (ID) with the distribution of the ratio healthy humans/elderly human (IV) resulted in a phosphate specific factor of 4.5 (3.3-6.7) (median; 25th - 75th percentile).


Assuntos
Fosfatos de Cálcio/toxicidade , Taxa de Filtração Glomerular/efeitos dos fármacos , Rim/efeitos dos fármacos , Nefrocalcinose/induzido quimicamente , Animais , Fosfatos de Cálcio/metabolismo , Taxa de Filtração Glomerular/fisiologia , Humanos , Rim/metabolismo , Nefrocalcinose/metabolismo , Nefrocalcinose/fisiopatologia , Ratos , Medição de Risco , Especificidade da Espécie
5.
Am J Physiol Renal Physiol ; 319(2): F202-F214, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32628541

RESUMO

Kidney stone disease is a crystal concretion formed in the kidneys that has been associated with an increased risk of chronic kidney disease. MicroRNAs are functionally involved in kidney injury. Data mining using a microRNA array database suggested that miR-21 may be associated with calcium oxalate monohydrate (COM)-induced renal tubular cell injury. Here, we confirmed that COM exposure significantly upregulated miR-21 expression, inhibited proliferation, promoted apoptosis, and caused lipid accumulation in an immortalized renal tubular cell line (HK-2). Moreover, inhibition of miR-21 enhanced proliferation and decreased apoptosis and lipid accumulation in HK-2 cells upon COM exposure. In a glyoxylate-induced mouse model of renal calcium oxalate deposition, increased miR-21 expression, lipid accumulation, and kidney injury were also observed. In silico analysis and subsequent experimental validation confirmed the peroxisome proliferator-activated receptor (PPAR)-α gene (PPARA) a key gene in fatty acid oxidation, as a direct miR-21 target. Suppression of miR-21 by miRNA antagomiR or activation of PPAR-α by its selective agonist fenofibrate significantly reduced renal lipid accumulation and protected against renal injury in vivo. In addition, miR-21 was significantly increased in urine samples from patients with calcium oxalate renal stones compared with healthy volunteers. In situ hybridization of biopsy samples from patients with nephrocalcinosis revealed that miR-21 was also significantly upregulated compared with normal kidney tissues from patients with renal cell carcinoma who underwent radical nephrectomy. These results suggested that miR-21 promoted calcium oxalate-induced renal tubular cell injury by targeting PPARA, indicating that miR-21 could be a potential therapeutic target and biomarker for nephrolithiasis.


Assuntos
Oxalato de Cálcio/farmacologia , Rim/lesões , MicroRNAs/farmacologia , PPAR alfa/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/genética , Biomarcadores/metabolismo , Oxalato de Cálcio/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Rim/metabolismo , Cálculos Renais/patologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , MicroRNAs/genética , Nefrocalcinose/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Mol Metab ; 40: 101020, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32439336

RESUMO

OBJECTIVE: Insulin signalling via phosphoinositide 3-kinase (PI3K) requires PIK3R1-encoded regulatory subunits. C-terminal PIK3R1 mutations cause SHORT syndrome, as well as lipodystrophy and insulin resistance (IR), surprisingly without fatty liver or metabolic dyslipidaemia. We sought to investigate this discordance. METHODS: The human pathogenic Pik3r1 Y657∗ mutation was knocked into mice by homologous recombination. Growth, body composition, bioenergetic and metabolic profiles were investigated on chow and high-fat diet (HFD). We examined adipose and liver histology, and assessed liver responses to fasting and refeeding transcriptomically. RESULTS: Like humans with SHORT syndrome, Pik3r1WT/Y657∗ mice were small with severe IR, and adipose expansion on HFD was markedly reduced. Also as in humans, plasma lipid concentrations were low, and insulin-stimulated hepatic lipogenesis was not increased despite hyperinsulinemia. At odds with lipodystrophy, however, no adipocyte hypertrophy nor adipose inflammation was found. Liver lipogenic gene expression was not significantly altered, and unbiased transcriptomics showed only minor changes, including evidence of reduced endoplasmic reticulum stress in the fed state and diminished Rictor-dependent transcription on fasting. Increased energy expenditure, which was not explained by hyperglycaemia nor intestinal malabsorption, provided an alternative explanation for the uncoupling of IR from dyslipidaemia. CONCLUSIONS: Pik3r1 dysfunction in mice phenocopies the IR and reduced adiposity without lipotoxicity of human SHORT syndrome. Decreased adiposity may not reflect bona fide lipodystrophy, but rather, increased energy expenditure, and we suggest that further study of brown adipose tissue in both humans and mice is warranted.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/genética , Transtornos do Crescimento/metabolismo , Hipercalcemia/metabolismo , Resistência à Insulina/genética , Doenças Metabólicas/metabolismo , Nefrocalcinose/metabolismo , Tecido Adiposo Marrom/metabolismo , Adiposidade , Animais , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Dieta Hiperlipídica , Dislipidemias/genética , Metabolismo Energético/genética , Fígado Gorduroso/metabolismo , Transtornos do Crescimento/genética , Hipercalcemia/genética , Inflamação/metabolismo , Insulina/metabolismo , Lipogênese , Fígado/metabolismo , Masculino , Doenças Metabólicas/genética , Camundongos , Camundongos Endogâmicos C57BL , Nefrocalcinose/genética , Obesidade/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo
7.
Sci Rep ; 10(1): 4864, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32184468

RESUMO

Excessive phosphorus intake adversely affects bone and mineral metabolism. Estrogen is one of the factors affecting fibroblast growth factor 23 (FGF23), a phosphorus-regulating hormone. However, the interaction between excess phosphorus and estrogen status has not been fully elucidated. This study investigated the involvement of estrogen in the effects of high phosphorus intake on bone metabolism and ectopic calcification in ovariectomized (OVX) rats. The interaction between high phosphorus diet and OVX was not observed in bone mineral density and aortic calcium. In contrast, high phosphorus intake markedly increased renal calcium concentration in sham rats, whereas the effect was attenuated in OVX rats, which was reversed by a selective estrogen-receptor modulator treatment. A strong positive correlation between renal calcium and serum FGF23 was observed. In addition, fibroblast growth factor receptor 1 (FGFR1: a predominant receptor of FGF23) inhibitor treatment partially decreased renal calcium concentrations in rats with high phosphorus intake. In conclusion, the effect of high phosphorus intake on bone metabolism and aortic calcification did not depend on the estrogen status; in contrast, high phosphorus intake synergistically induced nephrocalcinosis in the presence of estrogenic action on the bone. Furthermore, FGF23 was involved in the nephrocalcinosis induced by high phosphorus intake partially through FGFR1 signaling.


Assuntos
Estrogênios/metabolismo , Fatores de Crescimento de Fibroblastos/sangue , Nefrocalcinose/metabolismo , Fósforo/efeitos adversos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Aorta/metabolismo , Densidade Óssea/efeitos dos fármacos , Cálcio/metabolismo , Modelos Animais de Doenças , Feminino , Fatores de Crescimento de Fibroblastos/efeitos dos fármacos , Nefrocalcinose/sangue , Nefrocalcinose/induzido quimicamente , Ovariectomia/efeitos adversos , Pirimidinas/farmacologia , Cloridrato de Raloxifeno/farmacologia , Ratos
8.
EBioMedicine ; 50: 366-378, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31735555

RESUMO

BACKGROUND: Intrarenal calcium oxalate (CaOx) crystals induce inflammation and kidney tubular cell injury, which are processes that involve TLR4/NF-κB signalling. A recent genome-wide gene expression profile analysis of Randall's plaques in CaOx stone patients revealed that the expression of the long noncoding RNA H19 was significantly upregulated. However, to date, its role in kidney CaOx stones has not been reported. METHOD: A Gene Expression Omnibus (GEO) dataset was utilized to analyse gene expression profiles. Luciferase reporter, Western blotting, qRT-PCR, immunofluorescence staining and reactive oxygen species (ROS) assays were employed to study the molecular mechanism of HMGB1/TLR4/NF-κB regulation by H19 and miR-216b. In vitro and in vivo assays were performed to further confirm the proinflammatory and prooxidative stress effects. FINDING: H19 expression was significantly increased and positively correlated with the expression levels of HMGB1, TLR4 and NF-κB in Randall's plaques and glyoxylate-induced CaOx nephrocalcinosis mouse models. H19 interacted with miR-216b and suppressed its expression. Additionally, miR-216b inhibited HMGB1 expression by directly binding its 3'-untranslated region. Moreover, H19 downregulation inhibited HMGB1, TLR4 and NF-κB expression and suppressed CaOx nephrocalcinosis-induced renal tubular epithelial cell injury, NADPH oxidase, and oxidative stress in vivo and in vitro. Interestingly, miR-216b inhibition partially reversed the inhibitory effect of H19 knockdown on HMGB1 expression. INTERPRETATION: We determined that H19 might serve as a facilitator in the process of CaOx nephrocalcinosis-induced oxidative stress and renal tubular epithelial cell injury, and we revealed that the interaction between H19 and miR-216b could exert its effect via the HMGB1/TLR4/NF-κB pathway. FUNDING: This work was supported by the National Nature Science Foundation of China (Nos. 8196030190, 8190033175, 81370805, 81470935, 81900645, 81500534, and 81602236).


Assuntos
Oxalato de Cálcio/metabolismo , Células Epiteliais/metabolismo , Túbulos Renais/metabolismo , Nefrocalcinose/etiologia , Nefrocalcinose/metabolismo , RNA Longo não Codificante/genética , RNA não Traduzido/genética , Regiões 3' não Traduzidas , Animais , Biomarcadores , Oxalato de Cálcio/química , Linhagem Celular , Biologia Computacional , Modelos Animais de Doenças , Células Epiteliais/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteína HMGB1/genética , Humanos , Imuno-Histoquímica , Túbulos Renais/patologia , Masculino , Camundongos , Modelos Biológicos , Nefrocalcinose/patologia , Estresse Oxidativo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo
9.
J Pediatr ; 203: 391-399.e1, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30470382

RESUMO

OBJECTIVE: To determine whether multiple daily injections of parathyroid hormone (PTH) 1-34 are safe and effective as long-term therapy for children with hypoparathyroidism. STUDY DESIGN: Linear growth, bone accrual, renal function, and mineral homeostasis were studied in a long-term observational study of PTH 1-34 injection therapy in 14 children. METHODS: Subjects were 14 children with hypoparathyroidism attributable to autoimmune polyglandular syndrome type 1 (N = 5, ages 7-12 years) or calcium receptor mutation (N = 9, ages 7-16 years). Mean daily PTH 1-34 dose was 0.75 ± 0.15 µg/kg/day. Treatment duration was 6.9 ± 3.1 years (range 1.5-10 years). Patients were evaluated semiannually at the National Institutes of Health Clinical Center. RESULTS: Mean height velocity and lumbar spine, whole body, and femoral neck bone accretion velocities were normal throughout the study. In the first 2 years, distal one-third radius bone accrual velocity was reduced compared with normal children (P < .003). Serum alkaline phosphatase correlated with PTH 1-34 dose (P < .006) and remained normal (235.3 ± 104.8 [SD] U/L, N: 51-332 U/L). Mean serum and 24-hour urine calcium levels were 2.05 ± 0.11 mmol/L (N: 2.05-2.5 mmol/L) and 6.93 ± 1.3 mmol/24 hour (N: 1.25-7.5 mmol/24 hour), respectively-with fewer high urine calcium levels vs baseline during calcitriol and calcium treatment (P < .001). Nephrocalcinosis progressed in 5 of 12 subjects who had repeated renal imaging although renal function remained normal. CONCLUSIONS: Twice-daily or thrice-daily subcutaneous PTH 1-34 injections provided safe and effective replacement therapy for up to 10 years in children with hypoparathyroidism because of autoimmune polyglandular syndrome type 1 or calcium receptor mutation.


Assuntos
Estatura/efeitos dos fármacos , Hipoparatireoidismo/tratamento farmacológico , Hormônio Paratireóideo/uso terapêutico , Adolescente , Calcinose , Cálcio/sangue , Cálcio/urina , Criança , Creatinina/urina , Análise Mutacional de DNA , Feminino , Homeostase , Terapia de Reposição Hormonal , Humanos , Testes de Função Renal , Modelos Lineares , Masculino , Nefrocalcinose/metabolismo , Hormônio Paratireóideo/administração & dosagem , Fósforo/sangue , Fósforo/urina , Poliendocrinopatias Autoimunes/genética , Receptores de Detecção de Cálcio/genética , Resultado do Tratamento , Vitamina D/sangue
10.
Biosci Rep ; 38(1)2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29208768

RESUMO

Nephrocalcinosis involves the deposition of microscopic crystals in the tubular lumen or interstitium. While the clinical, biochemical, and genetic aspects of the diseases causing nephrocalcinosis have been elucidated, little is known about the cellular events in this calcification process. We previously reported a phenomenon involving the spontaneous formation of Ca2PO4 nodules in primary papillary renal cells from a patient with medullary nephrocalcinosis harboring a rare glial cell-derived neurotrophic factor (GDNF) gene variant. We also demonstrated that cultivating GDNF-silenced human kidney-2 (HK-2) cells in osteogenic conditions for 15 days triggered Ca2PO4 deposits. Given the reportedly close relationship between cell death and pathological calcification, aim of the present study was to investigate whether apoptosis is involved in the calcification of GDNF-silenced HK-2 cells under osteogenic conditions. Silenced and control cells were cultured in standard and osteogenic medium for 1, 5, and 15 days, and any Ca2PO4 deposition was identified by means of von Kossa staining and environmental SEM (ESEM) analyses. Based on the results of annexin V and propidium iodide (PI) analysis, and terminal deoxynucleotidyl transferase dUTP-biotin nick end labeling (TUNEL) assay, the silenced cells in the osteogenic medium showed a significant increase in the percentage of cells in the late phase of apoptosis and an increased Ca2PO4 deposition at 15 days. The results of quantitative real-time PCR (qRT-PCR) of BAX and BCL2, and in-cell Western analysis of caspases indicated that the cell death process was independent of caspase-3, -6, -7, and -9 activation, however. Using this model, we provide evidence of caspase-independent cell death triggering the calcification process in GDNF-silenced HK-2 cells.


Assuntos
Fosfatos de Cálcio/metabolismo , Morte Celular/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Nefrocalcinose/genética , Apoptose/genética , Caspases/genética , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Nefrocalcinose/metabolismo , Nefrocalcinose/patologia
11.
Morphologie ; 102(336): 48-53, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29102553

RESUMO

Primary hyperoxaluria is a rare genetic disorder characterized by oxalate overproduction, leading to kidney failure due to nephrocalcinosis, and is eventually responsible for systemic oxalosis. Bone impairment, secondary to oxalate deposits, is one of the many complications that may occur. Skeletal involvement can be difficult to diagnose because of lack of clinical symptoms and therefore needs to be confirmed by invasive testing, such as transiliac bone biopsy. If confirmed, bone oxalosis is the proof of disease severity and that combined liver-kidney transplantation should be performed.


Assuntos
Oxalato de Cálcio/metabolismo , Hiperoxalúria Primária/metabolismo , Ílio/patologia , Nefrocalcinose/metabolismo , Adulto , Biópsia , Densidade Óssea , Oxalato de Cálcio/urina , Humanos , Hiperoxalúria Primária/tratamento farmacológico , Hiperoxalúria Primária/genética , Hiperoxalúria Primária/urina , Ílio/citologia , Ílio/diagnóstico por imagem , Falência Renal Crônica/metabolismo , Falência Renal Crônica/terapia , Transplante de Rim , Masculino , Microrradiografia , Nefrocalcinose/diagnóstico por imagem , Nefrocalcinose/genética , Nefrocalcinose/urina , Osteoblastos/patologia , Piridoxina/uso terapêutico , Diálise Renal , Transaminases/genética
12.
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
13.
Hum Mol Genet ; 26(19): 3713-3721, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28934384

RESUMO

SHORT syndrome is a rare, recognizable syndrome resulting from heterozygous mutations in PIK3R1 encoding a regulatory subunit of phosphoinositide-3-kinase (PI3K). The condition is characterized by short stature, intrauterine growth restriction, lipoatrophy and a facial gestalt involving a triangular face, deep set eyes, low hanging columella and small chin. PIK3R1 mutations in SHORT syndrome result in reduced signaling through the PI3K-AKT-mTOR pathway. We performed whole exome sequencing for an individual with clinical features of SHORT syndrome but negative for PIK3R1 mutation and her parents. A rare de novo variant in PRKCE was identified. The gene encodes PKCε and, as such, the AKT-mTOR pathway function was assessed using phospho-specific antibodies with patient lymphoblasts and following ectopic expression of the mutant in HEK293 cells. Kinase analysis showed that the variant resulted in a partial loss-of-function. Whilst interaction with PDK1 and the mTORC2 complex component SIN1 was preserved in the mutant PKCε, it bound to SIN1 with a higher affinity than wild-type PKCε and the dynamics of mTORC2-dependent priming of mutant PKCε was altered. Further, mutant PKCε caused impaired mTORC2-dependent pAKT-S473 following rapamycin treatment. Reduced pFOXO1-S256 and pS6-S240/244 levels were also observed in the patient LCLs. To date, mutations in PIK3R1 causing impaired PI3K-dependent AKT activation are the only known cause of SHORT syndrome. We identify a SHORT syndrome child with a novel partial loss-of-function defect in PKCε. This variant causes impaired AKT activation via compromised mTORC2 complex function.


Assuntos
Transtornos do Crescimento/genética , Hipercalcemia/genética , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Doenças Metabólicas/genética , Nefrocalcinose/genética , Proteína Quinase C-épsilon/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Adolescente , Nanismo/genética , Feminino , Transtornos do Crescimento/metabolismo , Células HEK293 , Humanos , Hipercalcemia/metabolismo , Doenças Metabólicas/metabolismo , Mutação , Nefrocalcinose/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosforilação , Proteína Quinase C-épsilon/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
14.
Pediatr Endocrinol Rev ; 13(1): 468-76, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26540764

RESUMO

Diagnosis and management of pediatric nephrolithiasis/nephrocalcinosis is a very complex and challenging task for every pediatrician. It is based on correct. disease history taking, which may guide to the mode of inheritance (dominant, recessive, x-linked). Ethnicity and consanguinity should also be investigated since they predispose to high prevalence of certain disorders. One should always begin with cheap and available screening tests. Herein we will review clinical, biochemical, metabolic and genetic characteristics of the inherited diseases which lead to nephrolithiasis/nephrocalcinosis, such as: idiopathic hypercalciuria, renal hypophosphatemia, renal tubular acidosis, idiopathic infantile hypercalcemia, Dent disease, familial hypomagnesemia with hypercalciuria and nephrocalcinosis, hypocitraturia, cystinuria, primary hyperoxaluria and renal hypouricemia. Modern genetic techniques such as next generation sequencing enable nowadays diagnosis of rare disease using only a blood sample, trough massive parallel resequencing of many genes. This is very helpful for anuric patients or on dialysis where blood and urine biochemistry are not informative. Genetic testing also replaces invasive liver biopsy or unpleasant acidification tests and enables prenatal or early postnatal diagnosis.


Assuntos
Nefrocalcinose/genética , Nefrocalcinose/metabolismo , Nefrolitíase/genética , Nefrolitíase/metabolismo , Criança , Testes Genéticos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Nefropatias/complicações
15.
Kidney Int ; 88(1): 35-43, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25807034

RESUMO

The available publications on nephrocalcinosis are wide-ranging and have documented multiple causes and associations of macroscopic or radiological nephrocalcinosis, most often located in the renal medulla, with various metabolic and genetic disorders; in fact, so many and various are these that it is difficult to define a common underlying mechanism. We have reviewed nephrocalcinosis in relation to its definition, genetic associations, animal models, and putative mechanisms. We have concluded, and hypothesized, that nephrocalcinosis is primarily a renal interstitial process, resembling metastatic calcification, and that it may have some features in common with, and pathogenic links to, vascular calcification.


Assuntos
Cálcio/metabolismo , Nefrocalcinose/genética , Nefrocalcinose/metabolismo , Animais , Oxalato de Cálcio/metabolismo , Fosfatos de Cálcio/metabolismo , Modelos Animais de Doenças , Homeostase , Humanos , Túbulos Renais/metabolismo
16.
Nephrol Dial Transplant ; 30(6): 952-7, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25765846

RESUMO

BACKGROUND: Magnesium (Mg(2+)) is an essential ion for cell growth, neuroplasticity and muscle contraction. Blood Mg(2+) levels <0.7 mmol/L may cause a heterogeneous clinical phenotype, including muscle cramps and epilepsy and disturbances in K(+) and Ca(2+) homeostasis. Over the last decade, the genetic origin of several familial forms of hypomagnesaemia has been found. In 2000, mutations in FXYD2, encoding the γ-subunit of the Na(+)-K(+)-ATPase, were identified to cause isolated dominant hypomagnesaemia (IDH) in a large Dutch family suffering from hypomagnesaemia, hypocalciuria and chondrocalcinosis. However, no additional patients have been identified since then. METHODS: Here, two families with hypomagnesaemia and hypocalciuria were screened for mutations in the FXYD2 gene. Moreover, the patients were clinically and genetically characterized. RESULTS: We report a p.Gly41Arg FXYD2 mutation in two families with hypomagnesaemia and hypocalciuria. Interestingly, this is the same mutation as was described in the original study. As in the initial family, several patients suffered from muscle cramps, chondrocalcinosis and epilepsy. Haplotype analysis revealed an overlapping haplotype in all families, suggesting a founder effect. CONCLUSIONS: The recurrent p.Gly41Arg FXYD2 mutation in two new families with IDH confirms that FXYD2 mutation causes hypomagnesaemia. Until now, no other FXYD2 mutations have been reported which could indicate that other FXYD2 mutations will not cause hypomagnesaemia or are embryonically lethal.


Assuntos
Hipercalciúria/genética , Magnésio/sangue , Mutação/genética , Nefrocalcinose/genética , Erros Inatos do Transporte Tubular Renal/genética , ATPase Trocadora de Sódio-Potássio/genética , Adulto , Feminino , Genes Dominantes , Homeostase/genética , Humanos , Hipercalciúria/metabolismo , Masculino , Nefrocalcinose/metabolismo , Linhagem , Erros Inatos do Transporte Tubular Renal/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
17.
J Am Soc Nephrol ; 25(3): 574-86, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24204001

RESUMO

Mutations in PCBD1 are causative for transient neonatal hyperphenylalaninemia and primapterinuria (HPABH4D). Until now, HPABH4D has been regarded as a transient and benign neonatal syndrome without complications in adulthood. In our study of three adult patients with homozygous mutations in the PCBD1 gene, two patients were diagnosed with hypomagnesemia and renal Mg(2+) loss, and two patients developed diabetes with characteristics of maturity onset diabetes of the young (MODY), regardless of serum Mg(2+) levels. Our results suggest that these clinical findings are related to the function of PCBD1 as a dimerization cofactor for the transcription factor HNF1B. Mutations in the HNF1B gene have been shown to cause renal malformations, hypomagnesemia, and MODY. Gene expression studies combined with immunohistochemical analysis in the kidney showed that Pcbd1 is expressed in the distal convoluted tubule (DCT), where Pcbd1 transcript levels are upregulated by a low Mg(2+)-containing diet. Overexpression in a human kidney cell line showed that wild-type PCBD1 binds HNF1B to costimulate the FXYD2 promoter, the activity of which is instrumental in Mg(2+) reabsorption in the DCT. Of seven PCBD1 mutations previously reported in HPABH4D patients, five mutations caused proteolytic instability, leading to reduced FXYD2 promoter activity. Furthermore, cytosolic localization of PCBD1 increased when coexpressed with HNF1B mutants. Overall, our findings establish PCBD1 as a coactivator of the HNF1B-mediated transcription necessary for fine tuning FXYD2 transcription in the DCT and suggest that patients with HPABH4D should be monitored for previously unrecognized late complications, such as hypomagnesemia and MODY diabetes.


Assuntos
Diabetes Mellitus Tipo 2/etiologia , Fator 1-beta Nuclear de Hepatócito/metabolismo , Hidroliases/genética , Hipercalciúria/genética , Nefrocalcinose/genética , Erros Inatos do Transporte Tubular Renal/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Adolescente , Animais , Feminino , Células HEK293 , Humanos , Hidroliases/metabolismo , Hipercalciúria/metabolismo , Lactente , Túbulos Renais Distais/metabolismo , Magnésio/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Nefrocalcinose/metabolismo , Fenilcetonúrias/genética , Erros Inatos do Transporte Tubular Renal/metabolismo , Adulto Jovem
18.
PLoS Genet ; 9(2): e1003302, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23468644

RESUMO

Enamel-renal syndrome (ERS) is an autosomal recessive disorder characterized by severe enamel hypoplasia, failed tooth eruption, intrapulpal calcifications, enlarged gingiva, and nephrocalcinosis. Recently, mutations in FAM20A were reported to cause amelogenesis imperfecta and gingival fibromatosis syndrome (AIGFS), which closely resembles ERS except for the renal calcifications. We characterized three families with AIGFS and identified, in each case, recessive FAM20A mutations: family 1 (c.992G>A; g.63853G>A; p.Gly331Asp), family 2 (c.720-2A>G; g.62232A>G; p.Gln241_Arg271del), and family 3 (c.406C>T; g.50213C>T; p.Arg136* and c.1432C>T; g.68284C>T; p.Arg478*). Significantly, a kidney ultrasound of the family 2 proband revealed nephrocalcinosis, revising the diagnosis from AIGFS to ERS. By characterizing teeth extracted from the family 3 proband, we demonstrated that FAM20A(-/-) molars lacked true enamel, showed extensive crown and root resorption, hypercementosis, and partial replacement of resorbed mineral with bone or coalesced mineral spheres. Supported by the observation of severe ectopic calcifications in the kidneys of Fam20a null mice, we conclude that FAM20A, which has a kinase homology domain and localizes to the Golgi, is a putative Golgi kinase that plays a significant role in the regulation of biomineralization processes, and that mutations in FAM20A cause both AIGFS and ERS.


Assuntos
Amelogênese Imperfeita , Proteínas do Esmalte Dentário , Fibromatose Gengival , Nefrocalcinose , Amelogênese Imperfeita/diagnóstico , Amelogênese Imperfeita/genética , Amelogênese Imperfeita/metabolismo , Amelogênese Imperfeita/patologia , Animais , Calcinose/diagnóstico , Calcinose/genética , Calcinose/metabolismo , Esmalte Dentário/metabolismo , Esmalte Dentário/patologia , Proteínas do Esmalte Dentário/deficiência , Proteínas do Esmalte Dentário/genética , Proteínas do Esmalte Dentário/metabolismo , Fibromatose Gengival/diagnóstico , Fibromatose Gengival/genética , Fibromatose Gengival/patologia , Complexo de Golgi/metabolismo , Complexo de Golgi/patologia , Humanos , Rim/metabolismo , Rim/fisiopatologia , Camundongos , Mutação , Nefrocalcinose/diagnóstico , Nefrocalcinose/genética , Nefrocalcinose/metabolismo , Fosfotransferases/genética , Fosfotransferases/metabolismo
19.
J Oncol Pharm Pract ; 19(3): 254-6, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22948887

RESUMO

Hypomagnesemia is known to occur for a variety of renal, gastrointestinal and other causes, and is often associated with other electrolyte and metabolic disturbances. We present a case of isolated hypomagnesemia in a patient who had been treated with the chemotherapy agent capecitabine. The approach to diagnosis and treatment is discussed. We postulate that capecitabine may cause isolated hypomagnesemia, possibly due to renal magnesium loss.


Assuntos
Desoxicitidina/análogos & derivados , Fluoruracila/análogos & derivados , Hipercalciúria/induzido quimicamente , Nefrocalcinose/induzido quimicamente , Erros Inatos do Transporte Tubular Renal/induzido quimicamente , Capecitabina , Desoxicitidina/efeitos adversos , Desoxicitidina/uso terapêutico , Feminino , Fluoruracila/efeitos adversos , Fluoruracila/uso terapêutico , Humanos , Hipercalciúria/metabolismo , Magnésio/metabolismo , Pessoa de Meia-Idade , Nefrocalcinose/metabolismo , Erros Inatos do Transporte Tubular Renal/metabolismo
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA