Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Biol Chem ; 289(26): 18489-96, 2014 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-24828496

RESUMO

Epinephrine and norepinephrine are present in the pro-urine. ß-Adrenergic receptor (ß-AR) blockers administered to counteract sympathetic overstimulation in patients with congestive heart failure have a negative inotropic effect, resulting in reduced cardiac contractility. Positive inotropes, ß1-AR agonists, are used to improve cardiac functions. Active Ca(2+) reabsorption in the late distal convoluted and connecting tubules (DCT2/CNT) is initiated by Ca(2+) influx through the transient receptor potential vanilloid type 5 (TRPV5) Ca(2+) channel. Although it was reported that ß-ARs are present in the DCT2/CNT region, their role in active Ca(2+) reabsorption remains elusive. Here we revealed that ß1-AR, but not ß2-AR, is localized with TRPV5 in DCT2/CNT. Subsequently, treatment of TRPV5-expressing mouse DCT2/CNT primary cell cultures with the ß1-AR agonist dobutamine showed enhanced apical-to-basolateral transepithelial Ca(2+) transport. In human embryonic kidney (HEK293) cells, dobutamine was shown to stimulate cAMP production, signifying functional ß1-AR expression. Fura-2 experiments demonstrated increased activity of TRPV5 in response to dobutamine, which could be prevented by the PKA inhibitor H89. Moreover, nonphosphorylable T709A-TRPV5 and phosphorylation-mimicking T709D-TRPV5 mutants were unresponsive to dobutamine. Surface biotinylation showed that dobutamine did not affect plasma membrane abundance of TRPV5. In conclusion, activation of ß1-AR stimulates active Ca(2+) reabsorption in DCT2/CNT; an increase in TRPV5 activity via PKA phosphorylation of residue Thr-709 possibly plays an important role. These data explicate a calciotropic role in addition to the inotropic property of ß1-AR.


Assuntos
Canais de Cálcio/metabolismo , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/genética , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Humanos , Lactente , Camundongos , Camundongos Transgênicos , Receptores Adrenérgicos beta 1/genética , Transdução de Sinais , Canais de Cátion TRPV/genética
2.
Nephrol Dial Transplant ; 28(4): 879-89, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23136218

RESUMO

BACKGROUND: Cisplatin is an effective anti-neoplastic drug, but its clinical use is limited due to dose-dependent nephrotoxicity. The majority of cisplatin-treated patients develop hypomagnesaemia, often associated with a reduced glomerular filtration rate (GFR), polyuria and other electrolyte disturbances. The aim of this study is to unravel the molecular mechanism responsible for these particular electrolyte disturbances. METHODS: Two groups of 10 mice were injected intraperitoneally three times, once every 4 days, with cisplatin (5 mg/kg body weight,) or vehicle. Serum and urine electrolyte concentrations were determined. Next, renal mRNA levels of distal convoluted tubule (DCT) genes epithelial Mg(2+) channel TRPM6, the Na(+)-Cl(-) cotransporter (NCC), and parvalbumin (PV), as well as marker genes for other tubular segments were measured by real-time qPCR. Subsequently, renal protein levels of NCC, PV, aquaporin 1 and aquaporin 2 were determined using immunoblotting and immunohistochemistry (IHC). RESULTS: The cisplatin-treated mice developed significant polyuria (2.5 ± 0.3 and 0.9 ± 0.1 mL/24 h, cisplatin versus control, P < 0.05), reduced creatinine clearance rate (CCr) (0.18 ± 0.02 and 0.26 ± 0.02 mL/min, cisplatin versus control, P < 0.05) and a substantially reduced serum level of Mg(2+) (1.23 ± 0.03 and 1.58 ± 0.03 mmol/L, cisplatin versus control, P < 0.05), whereas serum Ca(2+), Na(+) and K(+) values were not altered. Measurements of 24 h urinary excretion demonstrated markedly increased Mg(2+), Ca(2+), Na(+) and K(+) levels in the cisplatin-treated group, whereas Pi levels were not changed. The mRNA levels of TRPM6, NCC and PV were significantly reduced in the cisplatin group. The expression levels of the marker genes for other tubular segments were unaltered, except for claudin-16, which was significantly up-regulated by the cisplatin treatment. The observed DCT-specific down-regulation was confirmed at the protein level. CONCLUSIONS: The present study identified the DCT as an important cisplatin-affected renal segment, explaining the high prevalence of hypomagnesaemia following treatment.


Assuntos
Antineoplásicos/toxicidade , Biomarcadores/metabolismo , Cisplatino/toxicidade , Nefropatias/complicações , Túbulos Renais Distais/efeitos dos fármacos , Deficiência de Magnésio/etiologia , Animais , Aquaporina 2/genética , Aquaporina 2/metabolismo , Western Blotting , Eletrólitos/metabolismo , Feminino , Taxa de Filtração Glomerular , Técnicas Imunoenzimáticas , Nefropatias/tratamento farmacológico , Nefropatias/patologia , Túbulos Renais Distais/lesões , Deficiência de Magnésio/diagnóstico , Deficiência de Magnésio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Parvalbuminas/genética , Parvalbuminas/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Droga/genética , Receptores de Droga/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Membro 3 da Família 12 de Carreador de Soluto , Simportadores/genética , Simportadores/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo
3.
Cell Mol Life Sci ; 69(6): 981-92, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21952826

RESUMO

Tissue transglutaminase (tTG) is a multifunctional Ca(2+)-dependent enzyme, catalyzing protein crosslinking. The transient receptor potential vanilloid (TRPV) family of cation channels was recently shown to contribute to the regulation of TG activities in keratinocytes and hence skin barrier formation. In kidney, where active transcellular Ca(2+) transport via TRPV5 predominates, the potential effect of tTG remains unknown. A multitude of factors regulate TRPV5, many secreted into the pro-urine and acting from the extracellular side. We detected tTG in mouse urine and in the apical medium of polarized cultures of rabbit connecting tubule and cortical collecting duct (CNT/CCD) cells. Extracellular application of tTG significantly reduced TRPV5 activity in human embryonic kidney cells transiently expressing the channel. Similarly, a strong inhibition of transepithelial Ca(2+) transport was observed after apical application of purified tTG to polarized rabbit CNT/CCD cells. Furthermore, tTG promoted the aggregation of the plasma membrane-associated fraction of TRPV5. Using patch clamp analysis, we observed a reduction in the pore diameter after tTG treatment, suggesting distinct structural changes in TRPV5 upon crosslinking by tTG. As N-linked glycosylation of TRPV5 is a key step in regulating channel function, we determined the effect of tTG in the N-glycosylation-deficient TRPV5 mutant. In the absence of N-linked glycosylation, TRPV5 was insensitive to tTG. Taken together, these observations imply that tTG is a novel extracellular enzyme inhibiting the activity of TRPV5. The inhibition of TRPV5 occurs in an N-glycosylation-dependent manner, signifying a common final pathway by which distinct extracellular factors regulate channel activity.


Assuntos
Cálcio/metabolismo , Canais de Cátion TRPV/fisiologia , Transglutaminases/fisiologia , Animais , Glicosilação , Células HEK293 , Humanos , Transporte de Íons , Coelhos , Canais de Cátion TRPV/antagonistas & inibidores
4.
J Am Soc Nephrol ; 23(11): 1824-34, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23024298

RESUMO

Urinary proteins that leak through the abnormal glomerulus in nephrotic syndrome may affect tubular transport by interacting with membrane transporters on the luminal side of tubular epithelial cells. Patients with nephrotic syndrome can develop nephrocalcinosis, which animal models suggest may develop from impaired transcellular Ca(2+) reabsorption via TRPV5 in the distal convoluted tubule (DCT). In nephrotic-range proteinuria, filtered plasminogen reaches the luminal side of DCT, where it is cleaved into active plasmin by urokinase. In this study, we found that plasmin purified from the urine of patients with nephrotic-range proteinuria inhibits Ca(2+) uptake in TRPV5-expressing human embryonic kidney 293 cells through the activation of protease-activated receptor-1 (PAR-1). Preincubation with a plasmin inhibitor, a PAR-1 antagonist, or a protein kinase C (PKC) inhibitor abolished the effect of plasmin on TRPV5. In addition, ablation of the PKC phosphorylation site S144 rendered TRPV5 resistant to the action of plasmin. Patch-clamp experiments showed that a decreased TRPV5 pore size and a reduced open probability accompany the plasmin-mediated reduction in Ca(2+) uptake. Furthermore, high-resolution nuclear magnetic resonance spectroscopy demonstrated specific interactions between calmodulin and residues 133-154 of the N-terminus of TRPV5 for both wild-type and phosphorylated (S144pS) peptides. In summary, PAR-1 activation by plasmin induces PKC-mediated phosphorylation of TRPV5, thereby altering calmodulin-TRPV5 binding, resulting in decreased channel activity. These results indicate that urinary plasmin could contribute to the downstream effects of proteinuria on the tubulointerstitium by negatively modulating TRPV5.


Assuntos
Fibrinolisina/farmacologia , Fibrinolisina/urina , Síndrome Nefrótica/urina , Proteinúria/urina , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Calmodulina/metabolismo , Células HEK293 , Humanos , Túbulos Renais Distais/efeitos dos fármacos , Túbulos Renais Distais/metabolismo , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Fosforilação , Proteína Quinase C/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor PAR-1/metabolismo , Serina/química , Canais de Cátion TRPV/química , Canais de Cátion TRPV/genética
5.
Nephrol Dial Transplant ; 27(11): 4061-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22778178

RESUMO

BACKGROUND: Klotho(-/-) mice display disturbed Ca(2+) and vitamin D homeostasis. Renal cytochrome p450 27b1 (Cyp27b1), the enzyme that catalyzes the hydrolysis to 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)), is increased in klotho(-/-) mice, and a 1,25(OH)(2)D(3)-deficient diet partially normalized Ca(2+) homeostasis in these klotho(-/-) mice. The aim of the present study was to further delineate the interplay between 1,25(OH)(2)D(3) and klotho and their relative contribution to the Ca(2+) homeostasis of klotho(-/-) mice. METHODS: Double-klotho(-/-)/Cyp27b1(-/-) mice were generated and mice aged 8-12 weeks were housed in metabolic cages to collect 24-h urine. Blood samples were taken and the animals were sacrificed, and the kidney and duodenum tissues were sampled for RNA extraction. The bone was fixed in 10% v/v formalin and analysed by microcomputed tomography (µCT) scans. RESULTS: Klotho(-/-)/Cyp27b1(-/-) mice, like Cyp27b1(-/-) mice, displayed significantly decreased serum total calcium concentrations compared with wild-type mice (1.44 ± 0.03 and 2.25 ± 0.02 mM) along with normal urinary total calcium excretion. Hyperphosphataemia of klotho(-/-) mice normalized to wild-type levels in klotho(-/-)/Cyp27b1(-/-) mice. The mRNA levels of duodenal transient receptor potential vanilloid subtype 6 (TRPV6) and calcium-binding protein-D(9K), and renal calbindin-D(28K) and NCX1 were significantly reduced in the double knockouts compared with wild-type or klotho(-/-) mice. Elevated TRPV5 protein levels in klotho(-/-) mice normalized to wild type in klotho(-/-)/Cyp27b1(-/-) mice, but were decreased in Cyp27b1(-/-) mice. µCT scans showed that klotho(-/-)/Cyp27b1(-/-) mice, as Cyp27b1(-/-) mice, display significant bone hypomineralization and severely decreased bone mass. Klotho(-/-) mice show a reduced bone mass and increased trabecular numbers. CONCLUSIONS: Klotho(-/-)/Cyp27b1(-/-) mice resemble Cyp27b1(-/-) mice. Since 1,25(OH)(2)D(3) is absent in these mice, our results imply that Ca(2+) homeostasis in klotho(-/-) mice is affected by their excessive 1,25(OH)(2)D(3) levels.


Assuntos
Osso e Ossos/metabolismo , Cálcio/sangue , Glucuronidase/metabolismo , Fosfatos/sangue , Deficiência de Vitamina D/sangue , Vitamina D/análogos & derivados , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Animais , Western Blotting , Cálcio/urina , Duodeno/metabolismo , Glucuronidase/genética , Homeostase , Imuno-Histoquímica , Proteínas Klotho , Camundongos , Camundongos Knockout , Fosfatos/urina , Reação em Cadeia da Polimerase em Tempo Real , Canais de Cátion TRPV/metabolismo , Vitamina D/sangue , Deficiência de Vitamina D/urina
6.
J Biol Chem ; 285(34): 26081-7, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20584906

RESUMO

Mg(2+) is an essential ion for many cellular processes, including protein synthesis, nucleic acid stability, and numerous enzymatic reactions. Mg(2+) homeostasis in mammals depends on the equilibrium between intestinal absorption, renal excretion, and exchange with bone. The transient receptor potential melastatin type 6 (TRPM6) is an epithelial Mg(2+) channel, which is abundantly expressed in the luminal membrane of the renal and intestinal cells. It functions as the gatekeeper of transepithelial Mg(2+) transport. Remarkably, TRPM6 combines a Mg(2+)-permeable channel with an alpha-kinase domain. Here, by the Ras recruitment system, we identified methionine sulfoxide reductase B1 (MsrB1) as an interacting protein of the TRPM6 alpha-kinase domain. Importantly, MsrB1 and TRPM6 are both present in the renal Mg(2+)-transporting distal convoluted tubules. MsrB1 has no effect on TRPM6 channel activity in the normoxic conditions. However, hydrogen peroxide (H(2)O(2)) decreased TRPM6 channel activity. Co-expression of MsrB1 with TRPM6 attenuated the inhibitory effect of H(2)O(2) (TRPM6, 67 +/- 5% of control; TRPM6 + MsrB1, 81 +/- 5% of control). Cell surface biotinylation assays showed that H(2)O(2) treatment does not affect the expression of TRPM6 at the plasma membrane. Next, mutation of Met(1755) to Ala in TRPM6 reduced the inhibitory effect of H(2)O(2) on TRPM6 channel activity (TRPM6 M1755A: 84 +/- 10% of control), thereby mimicking the action of MsrB1. Thus, these data suggest that MsrB1 recovers TRPM6 channel activity by reducing the oxidation of Met(1755) and could, thereby, function as a modulator of TRPM6 during oxidative stress.


Assuntos
Metionina Sulfóxido Redutases/fisiologia , Estresse Oxidativo , Canais de Cátion TRPM/metabolismo , Animais , Linhagem Celular , Humanos , Peróxido de Hidrogênio/farmacologia , Magnésio/metabolismo , Metionina/metabolismo , Camundongos , Mutação , Oxirredução , Canais de Cátion TRPM/genética
7.
Curr Biol ; 18(3): 168-76, 2008 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-18258429

RESUMO

BACKGROUND: The maintenance of the body's Mg(2+) balance is of great importance because of its involvement in numerous enzymatic systems and its intervention in neuromuscular excitability, protein synthesis, and nucleic acid stability. Recently, the transient receptor potential melastatin 6 (TRPM6) was identified as the gatekeeper of active Mg(2+) transport and therefore plays a crucial role in the regulation of Mg(2+) homeostasis. Remarkably, TRPM6 combines a Mg(2+) channel with an alpha-kinase domain whose function remains elusive. RESULTS: Here, we identify the receptor for activated C-kinase 1 (RACK1) as the first regulatory protein of TRPM6 that associates with the alpha-kinase domain. RACK1 and TRPM6 are both present in renal Mg(2+)-transporting distal convoluted tubules. We demonstrate that RACK1 inhibits channel activity in an alpha-kinase activity-dependent manner, whereas small interference (si) RNA-mediated knockdown of RACK1 increases the current. Moreover, threonine(1851) in the alpha-kinase domain was identified as an autophosphorylation site of which the phosphorylation state is essential for the inhibitory effect of RACK1. Importantly, threonine(1851) was crucial for the Mg(2+) sensitivity of TRPM6 autophosphorylation and channel activity. TRPM6 channel activity was less sensitive to Mg(2+) when RACK1 was knocked down by siRNA. Finally, activation of protein kinase C by phorbol 12-myristate 13-acetate-PMA prohibited the inhibitory effect of RACK1 on TRPM6 channel activity. CONCLUSIONS: We propose a unique mode of TRPM6 regulation in which the Mg(2+) influx is controlled by RACK1 through its interaction with the alpha-kinase and the phosphorylation state of the threonine(1851) residue.


Assuntos
Fosfotransferases/metabolismo , Receptores de Superfície Celular/metabolismo , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Membrana Celular/enzimologia , Membrana Celular/metabolismo , Regulação da Expressão Gênica , Biblioteca Gênica , Humanos , Rim/metabolismo , Camundongos , Fosforilação , Ligação Proteica , Proteína Quinase C/metabolismo , Estrutura Terciária de Proteína , Receptores de Quinase C Ativada , Receptores de Superfície Celular/genética , Canais de Cátion TRPM/química , Canais de Cátion TRPM/genética , Técnicas do Sistema de Duplo-Híbrido
8.
Nephron Physiol ; 117(2): p11-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20814221

RESUMO

BACKGROUND: Hypomagnesemia with secondary hypocalcemia is due to disturbed renal and intestinal magnesium (Mg(2+)) (re)absorption. The underlying defect is a mutation in the transient receptor potential melastatin type 6 (TRPM6), a Mg(2+)-permeable ion channel expressed in the kidney and intestine. Our aim was to characterize homozygous (-/-) and heterozygous (+/-) TRPM6 knockout mice with respect to Mg(2+) homeostasis. METHODS: TRPM6(+/-) mice were bred on a normal (0.19% wt/wt Mg(2+)) and high (0.48% wt/wt Mg(2+)) Mg(2+) diet. In the offspring, 24-hour urinary Mg(2+) and calcium excretion as well as serum concentrations of both were determined. TRPM6 mRNA expression in the kidney and colon was measured. RESULTS: On the regular diet, 30% of the offspring were TRPM6 wild-type ((+/+)), 70% were TRPM6(+/-), and none were TRPM6(-/-). The genotypic distribution of the litters remained the same on the 0.48% Mg(2+) diet. In TRPM6(+/-) mice on both diets, serum Mg(2+) levels were significantly lower, and renal and intestinal TRPM6 mRNA expression was reduced. Urinary Mg(2+) excretion was unaffected. CONCLUSIONS: Homozygous TRPM6 deletion is embryonic lethal in mice. Heterozygous deletion of TRPM6 results in a mild hypomagnesemia. The Mg(2+)-enriched diet could not compensate for either embryonic lethality or hypomagnesemia caused by TRPM6 deficiency.


Assuntos
Deleção de Genes , Genes Letais , Deficiência de Magnésio/genética , Canais de Cátion TRPM/genética , Animais , Cálcio/urina , Fezes/química , Feminino , Expressão Gênica/fisiologia , Genótipo , Heterozigoto , Magnésio/sangue , Magnésio/urina , Deficiência de Magnésio/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Índice de Gravidade de Doença , Canais de Cátion TRPM/metabolismo
9.
J Am Soc Nephrol ; 20(8): 1705-13, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19470676

RESUMO

Hypercalciuria increases the risk for urolithiasis, but renal adaptive mechanisms reduce this risk. For example, transient receptor potential vanilloid 5 knockout (TPRV5(-/-)) mice lack kidney stones despite urinary calcium (Ca(2+)) wasting and hyperphosphaturia, perhaps as a result of their significant polyuria and urinary acidification. Here, we investigated the mechanisms linking hypercalciuria with these adaptive mechanisms. Exposure of dissected mouse outer medullary collecting ducts to high (5.0 mM) extracellular Ca(2+) stimulated H(+)-ATPase activity. In TRPV5(-/-) mice, activation of the renal Ca(2+)-sensing receptor promoted H(+)-ATPase-mediated H(+) excretion and downregulation of aquaporin 2, leading to urinary acidification and polyuria, respectively. Gene ablation of the collecting duct-specific B1 subunit of H(+)-ATPase in TRPV5(-/-) mice abolished the enhanced urinary acidification, which resulted in severe tubular precipitations of Ca(2+)-phosphate in the renal medulla. In conclusion, activation of Ca(2+)-sensing receptor by increased luminal Ca(2+) leads to urinary acidification and polyuria. These beneficial adaptations facilitate the excretion of large amounts of soluble Ca(2+), which is crucial to prevent the formation of kidney stones.


Assuntos
Hipercalciúria/urina , Nefrolitíase/urina , Receptores de Detecção de Cálcio/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Aquaporina 2/metabolismo , Cálcio/urina , Canais de Cálcio/genética , Concentração de Íons de Hidrogênio , Mucosa Intestinal/metabolismo , Rim/metabolismo , Túbulos Renais Coletores/enzimologia , Camundongos , Camundongos Knockout , Fenótipo , Proteínas de Transporte de Fosfato/metabolismo , Canais de Cátion TRPV/genética , ATPases Vacuolares Próton-Translocadoras/genética
10.
J Clin Invest ; 115(6): 1651-8, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15902302

RESUMO

Thiazide diuretics enhance renal Na+ excretion by blocking the Na+-Cl- cotransporter (NCC), and mutations in NCC result in Gitelman syndrome. The mechanisms underlying the accompanying hypocalciuria and hypomagnesemia remain debated. Here, we show that enhanced passive Ca2+ transport in the proximal tubule rather than active Ca2+ transport in distal convolution explains thiazide-induced hypocalciuria. First, micropuncture experiments in mice demonstrated increased reabsorption of Na+ and Ca2+ in the proximal tubule during chronic hydrochlorothiazide (HCTZ) treatment, whereas Ca2+ reabsorption in distal convolution appeared unaffected. Second, HCTZ administration still induced hypocalciuria in transient receptor potential channel subfamily V, member 5-knockout (Trpv5-knockout) mice, in which active distal Ca2+ reabsorption is abolished due to inactivation of the epithelial Ca2+ channel Trpv5. Third, HCTZ upregulated the Na+/H+ exchanger, responsible for the majority of Na+ and, consequently, Ca2+ reabsorption in the proximal tubule, while the expression of proteins involved in active Ca2+ transport was unaltered. Fourth, experiments addressing the time-dependent effect of a single dose of HCTZ showed that the development of hypocalciuria parallels a compensatory increase in Na+ reabsorption secondary to an initial natriuresis. Hypomagnesemia developed during chronic HCTZ administration and in NCC-knockout mice, an animal model of Gitelman syndrome, accompanied by downregulation of the epithelial Mg2+ channel transient receptor potential channel subfamily M, member 6 (Trpm6). Thus, Trpm6 downregulation may represent a general mechanism involved in the pathogenesis of hypomagnesemia accompanying NCC inhibition or inactivation.


Assuntos
Cálcio/metabolismo , Túbulos Renais Proximais/metabolismo , Magnésio/metabolismo , Erros Inatos do Transporte Tubular Renal/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Desequilíbrio Hidroeletrolítico/metabolismo , Animais , Benzotiadiazinas , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Distúrbios do Metabolismo do Cálcio/induzido quimicamente , Distúrbios do Metabolismo do Cálcio/metabolismo , Distúrbios do Metabolismo do Cálcio/patologia , Modelos Animais de Doenças , Diuréticos , Regulação para Baixo , Humanos , Transporte de Íons/genética , Túbulos Renais Proximais/patologia , Camundongos , Camundongos Knockout , Erros Inatos do Transporte Tubular Renal/genética , Erros Inatos do Transporte Tubular Renal/patologia , Inibidores de Simportadores de Cloreto de Sódio/toxicidade , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/genética , Canais de Cátion TRPV , Regulação para Cima , Desequilíbrio Hidroeletrolítico/induzido quimicamente , Desequilíbrio Hidroeletrolítico/patologia
11.
J Clin Invest ; 112(12): 1906-14, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14679186

RESUMO

Ca2+ ions play a fundamental role in many cellular processes, and the extracellular concentration of Ca2+ is kept under strict control to allow the proper physiological functions to take place. The kidney, small intestine, and bone determine the Ca2+ flux to the extracellular Ca2+ pool in a concerted fashion. Transient receptor potential (TRP) cation channel subfamily V, members 5 and 6 (TRPV5 and TRPV6) have recently been postulated to be the molecular gatekeepers facilitating Ca2+ influx in these tissues and are members of the TRP family, which mediates diverse biological effects ranging from pain perception to male aggression. Genetic ablation of TRPV5 in the mouse allowed us to investigate the function of this novel Ca2+ channel in maintaining the Ca2+ balance. Here, we demonstrate that mice lacking TRPV5 display diminished active Ca2+ reabsorption despite enhanced vitamin D levels, causing severe hypercalciuria. In vivo micropuncture experiments demonstrated that Ca2+ reabsorption was malfunctioning within the early part of the distal convolution, exactly where TRPV5 is localized. In addition, compensatory hyperabsorption of dietary Ca2+ was measured in TRPV5 knockout mice. Furthermore, the knockout mice exhibited significant disturbances in bone structure, including reduced trabecular and cortical bone thickness. These data demonstrate the key function of TRPV5 in active Ca2+ reabsorption and its essential role in the Ca2+ homeostasis.


Assuntos
Osso e Ossos/fisiologia , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Canais de Cálcio/fisiologia , Cálcio/metabolismo , Rim/metabolismo , Absorção , Animais , Sinalização do Cálcio , Cátions , Feminino , Fêmur/metabolismo , Biblioteca Gênica , Genótipo , Imuno-Histoquímica , Íons , Nefropatias/metabolismo , Masculino , Camundongos , Camundongos Knockout , Modelos Genéticos , Osteoporose , Fenótipo , Potássio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sódio/metabolismo , Canais de Cátion TRPV , Fatores de Tempo
12.
Oncotarget ; 7(18): 24928-41, 2016 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27102152

RESUMO

Trpv5 plays an important role in calcium (Ca2+) homeostasis, among others by mediating renal calcium reabsorption. Accordingly, Trpv5 deficiency strongly stresses Ca2+ homeostasis in order to maintain stable serum Ca2+. We addressed the impact of lifelong challenge of calcium homeostasis on the bone phenotype of these mice.Aging significantly increased serum 1,25(OH)2D3 and PTH levels in both genotypes but they were more elevated in Trpv5-/- mice, whereas serum Ca2+ was not affected by age or genotype. Age-related changes in trabecular and cortical bone mass were accelerated in Trpv5-/- mice, including reduced trabecular and cortical bone thickness as well as reduced bone mineralization. No effect of Trpv5 deficiency on bone strength was observed. In 78-week-old mice no differences were observed between the genotypes regarding urinary deoxypyridinoline, osteoclast number, differentiation and activity as well as osteoclast precursor numbers, as assessed by flow cytometry.In conclusion, life-long challenge of Ca2+ homeostasis present in Trpv5-/- mice causes accelerated bone aging and a low cortical and trabecular bone mass phenotype. The phenotype of the Trpv5-/- mice suggests that maintenance of adequate circulatory Ca2+ levels in patients with disturbances in Ca2+ homeostasis should be a priority in order to prevent bone loss at older age.


Assuntos
Envelhecimento/metabolismo , Osso e Ossos/metabolismo , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Homeostase/fisiologia , Canais de Cátion TRPV/metabolismo , Animais , Densidade Óssea/fisiologia , Osso e Ossos/patologia , Calcificação Fisiológica/fisiologia , Canais de Cálcio/deficiência , Masculino , Camundongos , Camundongos Knockout , Canais de Cátion TRPV/deficiência
13.
FASEB J ; 16(11): 1398-406, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12205031

RESUMO

Pseudovitamin D-deficiency rickets (PDDR) is an autosomal disease characterized by hyperparathyroidism, rickets, and undetectable levels of 1,25-dihydroxyvitaminD3 (1,25(OH)2D3). Mice in which the 25-hydroxyvitamin D3-1alpha-hydroxylase (1alpha-OHase) gene was inactivated presented the same clinical phenotype as patients with PDDR and were used to study renal expression of the epithelial Ca2+ channel (ECaC1), the calbindins, Na+/Ca2+ exchanger (NCX1), and Ca2+-ATPase (PMCA1b). Serum Ca2+ (1.20+/-0.05 mM) and mRNA/protein expression of ECaC1 (41+/-3%), calbindin-D28K (31+/-2%), calbindin-D9K (58+/-7%), NCX1 (10+/-2%), PMCA1b (96+/-4%) were decreased in 1alpha-OHase-/- mice compared with 1alpha-OHase+/- littermates. Feeding these mice a Ca2+-enriched diet normalized serum Ca2+ levels and expression of Ca2+ proteins except for calbindin-D9K expression. 1,25(OH)2D3 repletion resulted in increased expression of Ca2+ transport proteins and normalization of serum Ca2+ levels. Localization of Ca2+ transport proteins was clearly polarized in which ECaC1 was localized along the apical membrane, calbindin-D28K in the cytoplasm, and calbindin-D9K along the apical and basolateral membranes, resulting in a comprehensive mechanism facilitating renal transcellular Ca2+ transport. This study demonstrated that high dietary Ca2+ intake is an important regulator of the renal Ca2+ transport proteins in 1,25(OH)2D3-deficient status and thus contributes to the normalization of blood Ca2+ levels.


Assuntos
25-Hidroxivitamina D3 1-alfa-Hidroxilase/genética , Calcitriol/farmacologia , Proteínas de Ligação ao Cálcio/biossíntese , Cálcio/farmacologia , Rim/metabolismo , Administração Oral , Animais , Calbindina 1 , Calbindinas , Calcitriol/administração & dosagem , Cálcio/administração & dosagem , Cálcio/sangue , Canais de Cálcio/análise , Proteínas de Ligação ao Cálcio/análise , Proteínas de Ligação ao Cálcio/genética , ATPases Transportadoras de Cálcio/biossíntese , ATPases Transportadoras de Cálcio/genética , Proteínas de Transporte de Cátions , Rim/química , Camundongos , Camundongos Knockout , ATPases Transportadoras de Cálcio da Membrana Plasmática , RNA Mensageiro/biossíntese , Raquitismo/genética , Raquitismo/metabolismo , Proteína G de Ligação ao Cálcio S100/biossíntese , Proteína G de Ligação ao Cálcio S100/genética , Trocador de Sódio e Cálcio/biossíntese , Trocador de Sódio e Cálcio/genética , Canais de Cátion TRPV , Regulação para Cima , Deficiência de Vitamina D/genética , Deficiência de Vitamina D/metabolismo
14.
Clin Kidney J ; 5(6): 535-44, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26069797

RESUMO

BACKGROUND: Furosemide is a loop diuretic, which blocks the Na(+), K(+), 2Cl(-) cotransporter (NKCC2) in the thick ascending limb of Henle (TAL). By diminishing sodium (Na(+)) reabsorption, loop diuretics reduce the lumen-positive transepithelial voltage and consequently diminish paracellular transport of magnesium (Mg(2+)) and calcium (Ca(2+)) in TAL. Indeed, furosemide promotes urinary Mg(2+) excretion; however, it is unclear whether this leads, especially during prolonged treatment, to hypomagnesaemia. The aim of the present study was, therefore, to determine the effect of chronic furosemide application on renal Mg(2+) handling in mice. METHODS: Two groups of 10 mice received an osmotic minipump subcutaneously for 7 days with vehicle or 30 mg/kg/day furosemide. Serum and urine electrolyte concentrations were determined. Next, renal mRNA levels of the epithelial Mg(2+) channel (TRPM6), the Na(+), Cl(-) cotransporter (NCC), the epithelial Ca(2+) channel (TRPV5), the cytosolic Ca(2+)-binding protein calbindin-D28K, as well parvalbumin (PV), claudin-7 (CLDN7) and claudin-8 (CLDN8), the epithelial Na(+) channel (ENaC) and the Na(+)-H(+) exchanger 3 (NHE3) were determined by real-time quantitative polymerase chain reaction. Renal protein levels of NCC, TRPV5, calbindin-D28K and ENaC were also measured using semi-quantitative immunohistochemistry and immunoblotting. RESULTS: The mice chronically treated with 30 mg/kg/day furosemide displayed a significant polyuria (2.1 ± 0.3 and 1.3 ± 0.2 mL/24 h, furosemide versus control respectively, P < 0.05). Furosemide treatment resulted in increased serum concentrations of Na(+) [158 ± 3 (treated) and 147 ± 1 mmol/L (control), P < 0.01], whereas serum K(+), Ca(2+) and Mg(2+) values were not significantly altered in mice treated with furosemide. Urinary excretion of Na(+), K(+), Ca(2+) and Mg(2+) was not affected by chronic furosemide treatment. The present study shows specific renal upregulation of TRPM6, NCC, TRPV5 and calbindin-D28K. CONCLUSIONS: During chronic furosemide treatment, enhanced active reabsorption of Mg(2+) via the epithelial channel TRPM6 in DCT compensates for the reduced reabsorption of Mg(2+) in TAL.

15.
J Biol Chem ; 284(22): 14788-95, 2009 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-19329436

RESUMO

The maintenance of the Mg(2+) balance of the body is essential for neuromuscular excitability, protein synthesis, nucleic acid stability, and numerous enzymatic systems. The Transient Receptor Potential Melastatin 6 (TRPM6) functions as the gatekeeper of transepithelial Mg(2+) transport. However, the molecular regulation of TRPM6 channel activity remains elusive. Here, we identified the repressor of estrogen receptor activity (REA) as an interacting protein of TRPM6 that binds to the 6(th), 7(th), and 8(th) beta-sheets in its alpha-kinase domain. Importantly, REA and TRPM6 are coexpressed in renal Mg(2+)-transporting distal convoluted tubules (DCT). We demonstrated that REA significantly inhibits TRPM6, but not its closest homologue TRPM7, channel activity. This inhibition occurs in a phosphorylation-dependent manner, since REA has no effect on the TRPM6 phosphotransferase-deficient mutant (K1804R), while it still binds to this mutant. Moreover, activation of protein kinase C by phorbol 12-myristate 13-acetate-PMA potentiated the inhibitory effect of REA on TRPM6 channel activity. Finally, we showed that the interaction between REA and TRPM6 is a dynamic process, as short-term 17beta-estradiol treatment disassociates the binding between these proteins. In agreement with this, 17beta-estradiol treatment significantly stimulates the TRPM6-mediated current in HEK293 cells. These results suggest a rapid pathway for the effect of estrogen on Mg(2+) homeostasis in addition to its transcriptional effect. Together, these data indicate that REA operates as a negative feedback modulator of TRPM6 in the regulation of active Mg(2+) (re)absorption and provides new insight into the molecular mechanism of renal transepithelial Mg(2+) transport.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Estradiol/farmacologia , Proteínas Repressoras/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Rim/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Proibitinas , Ligação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/química
16.
J Clin Invest ; 119(4): 936-42, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19307729

RESUMO

Primary hypomagnesemia is a heterogeneous group of disorders characterized by renal or intestinal magnesium (Mg2+) wasting, resulting in tetany, cardiac arrhythmias, and seizures. The kidney plays an essential role in maintaining blood Mg2+ levels, with a prominent function for the Mg2+-transporting channel transient receptor potential cation channel, subfamily M, member 6 (TRPM6) in the distal convoluted tubule (DCT). In the DCT, Mg2+ reabsorption is an active transport process primarily driven by the negative potential across the luminal membrane. Here, we studied a family with isolated autosomal dominant hypomagnesemia and used a positional cloning approach to identify an N255D mutation in KCNA1, a gene encoding the voltage-gated potassium (K+) channel Kv1.1. Kv1.1 was found to be expressed in the kidney, where it colocalized with TRPM6 along the luminal membrane of the DCT. Upon overexpression in a human kidney cell line, patch clamp analysis revealed that the KCNA1 N255D mutation resulted in a nonfunctional channel, with a dominant negative effect on wild-type Kv1.1 channel function. These data suggest that Kv1.1 is a renal K+ channel that establishes a favorable luminal membrane potential in DCT cells to control TRPM6-mediated Mg2+ reabsorption.


Assuntos
Canal de Potássio Kv1.1/genética , Deficiência de Magnésio/genética , Mutação de Sentido Incorreto , Sequência de Aminoácidos , Sequência de Bases , Brasil , Linhagem Celular , Mapeamento Cromossômico , Análise Mutacional de DNA , Feminino , Genes Dominantes , Humanos , Rim/metabolismo , Canal de Potássio Kv1.1/química , Canal de Potássio Kv1.1/metabolismo , Deficiência de Magnésio/metabolismo , Masculino , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Linhagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Canais de Cátion TRPM/metabolismo , Transfecção
17.
Pflugers Arch ; 457(1): 91-101, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18392847

RESUMO

The transient receptor potential vanilloid channels 5 and 6 (TRPV5/6) are the most Ca(2+)-selective channels within the TRP superfamily of ion channels. These epithelial Ca(2+) channels are regulated at different intra- and extracellular sites by the feedback response of Ca(2+) itself, calciotropic hormones, and by TRPV5/6-associated proteins. In the present study, bioinformatics was used to search for novel TRPV5/6-associated genes. By including pull-down assays and functional analysis, Nipsnap1-a hitherto functionally uncharacterized globular protein-was identified as a novel factor involved in the regulation of TRPV6. Electrophysiological recordings revealed that Nipsnap1 abolishes TRPV6 currents. Subsequent biotinylation assays showed that TRPV6 plasma membrane expression did not change in the presence of Nipsnap1, suggesting that TRPV6 inhibition by Nipsnap1 is independently regulated from reduced cell surface channel expression. In addition, semi-quantitative reverse transcriptase PCR and immunohistochemical labeling of Nipsnap1 indicated that Nipsnap1 is expressed in mouse intestinal tissues-where TRPV6 is predominantly expressed-but that it does not co-localize with TRPV5 in the kidney. In conclusion, this study presents the first physiological function of Nipsnap1 as an associated protein inhibiting TRPV6 activity that possibly exerts its effect directly at the plasma membrane.


Assuntos
Canais de Cálcio/fisiologia , Proteínas/fisiologia , Canais de Cátion TRPV/fisiologia , Sequência de Aminoácidos , Canais de Cálcio/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Biologia Computacional , Eletrofisiologia , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Ligação Proteica , Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Cátion TRPV/genética , Distribuição Tecidual
18.
J Am Soc Nephrol ; 17(11): 3020-7, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17005931

RESUMO

The epithelial Ca(2+) channel TRPV5 facilitates apical Ca(2+) entry during active Ca(2+) reabsorption in the distal convoluted tubule. In this process, cytosolic Ca(2+) remains at low nontoxic concentrations because the Ca(2+) influx is buffered rapidly by calbindin-D(28K). Subsequently, Ca(2+) that is bound to calbindin-D(28K) is shuttled toward the basolateral Ca(2+) extrusion systems. For addressing the in vivo role of TRPV5 and calbindin-D(28K) in the maintenance of the Ca(2+) balance, single- and double-knockout mice of TRPV5 and calbindin-D(28K) (TRPV5(-/-), calbindin-D(28K)(-/-), and TRPV5(-/-)/calbindin-D(28K)(-/-)) were characterized. These mice strains were fed two Ca(2+) diets (0.02 and 2% wt/wt) to investigate the influence of dietary Ca(2+) content on the Ca(2+) balance. Urine analysis indicated that TRPV5(-/-)/calbindin-D(28K)(-/-) mice exhibit on both diets hypercalciuria compared with wild-type mice. Ca(2+) excretion in TRPV5(-/-)/calbindin-D(28K)(-/-) mice was not significantly different from TRPV5(-/-) mice, whereas calbindin-D(28K)(-/-) mice did not show hypercalciuria. The similarity between TRPV5(-/-)/calbindin-D(28K)(-/-) and TRPV5(-/-) mice was supported further by an equivalent increase in renal calbindin-D(9K) expression and in intestinal Ca(2+) hyperabsorption as a result of upregulation of calbindin-D(9K) and TRPV6 expression in the duodenum. Elevated serum parathyroid hormone and 1,25-dihydroxyvitamin D(3) levels accompanied the enhanced expression of the Ca(2+) transporters. Intestinal Ca(2+) absorption and expression of calbindin-D(9K) and TRPV6, as well as serum parameters of the calbindin-D(28K)(-/-) mice, did not differ from those of wild-type mice. These results underline the gatekeeper function of TRPV5 being the rate-limiting step in active Ca(2+) reabsorption, unlike calbindin-D(28K), which possibly is compensated by calbindin-D(9K).


Assuntos
Canais de Cálcio/fisiologia , Cálcio/metabolismo , Proteína G de Ligação ao Cálcio S100/genética , Canais de Cátion TRPV/fisiologia , Absorção , Animais , Calbindina 1 , Calbindinas , Canais de Cálcio/genética , Camundongos , Camundongos Knockout , Canais de Cátion TRPV/genética
19.
J Am Soc Nephrol ; 17(1): 26-30, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16380433

RESUMO

Transient receptor potential vallinoid 5 (TRPV5) and TRPV6 are the most Ca2+-selective members of the TRP superfamily and are essential for active Ca2+ (re)absorption in epithelia. However, little is known about intracellular proteins that regulate the activity of these channels. This study identified BSPRY (B-box and SPRY-domain containing protein) as a novel factor involved in the control of TRPV5. The interaction between BSPRY and TRPV5 by GST pull-down and co-immunoprecipitation assays was demonstrated. BSPRY showed co-localization with TRPV5 in mouse kidney. Expression of BSPRY resulted in a significant reduction of the Ca2+ influx in Madin-Darby Canine Kidney cells that stably express TRPV5 without affecting channel cell-surface abundance. Finally, BSPRY expression in kidney was increased in 25-hydroxyvitamin D3-1alpha-hydroxylase knockout mice, suggesting an inverse regulation by vitamin D3. Together, these results demonstrate the physiologic role of the novel protein BSPRY in the regulation of epithelial Ca2+ transport via negative modulation of TRPV5 activity.


Assuntos
Proteínas/fisiologia , Canais de Cátion TRPV/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Calcitriol/fisiologia , Cálcio/metabolismo , Canais de Cálcio/análise , Canais de Cálcio/fisiologia , Linhagem Celular , Dados de Sequência Molecular , Proteínas/análise , Proteínas/química , Canais de Cátion TRPV/análise , Canais de Cátion TRPV/fisiologia , Xenopus
20.
Am J Physiol Renal Physiol ; 291(6): F1177-83, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16705151

RESUMO

Aging is associated with alterations in Ca2+ homeostasis, which predisposes elder people to hyperparathyroidism and osteoporosis. Intestinal Ca2+ absorption decreases with aging and, in particular, active transport of Ca2+ by the duodenum. In addition, there are age-related changes in renal Ca2+ handling. To examine age-related changes in expression of the renal and intestinal epithelial Ca2+ channels, control (TRPV5+/+) and TRPV5 knockout (TRPV5-/-) mice aged 10, 30, and 52 wk were studied. Aging of TRPV5(+/+) mice resulted in a tendency toward increased renal Ca2+ excretion and significantly decreased intestinal Ca2+ absorption, which was accompanied by reduced expression of TRPV5 and TRPV6, respectively, despite increased serum 1,25(OH)2D3 levels. Similarly, in TRPV5-/- mice the existing renal Ca2+ loss was more pronounced in elder animals, whereas the compensatory intestinal Ca2+ absorption and TRPV6 expression declined with aging. In both mice strains, aging resulted in a resistance to 1,25(OH)2D3 and diminished renal vitamin D receptor mRNA levels, whereas serum Ca2+ levels remained constant. Furthermore, 52-wk-old TRPV5-/- mice showed severe hyperparathyroidism, whereas PTH levels in elder TRPV5+/+ mice remained normal. In 52-wk-old TRPV5-/- mice, serum osteocalcin levels were increased in accordance with the elevated PTH levels, suggesting an increased bone turnover in these mice. In conclusion, downregulation of TRPV5 and TRPV6 is likely involved in the impaired Ca2+ (re)absorption during aging. Moreover, TRPV5-/- mice likely develop age-related hyperparathyroidism and osteoporotic characteristics before TRPV5+/+ mice, demonstrating the importance of the epithelial Ca2+ channels in Ca2+ homeostasis.


Assuntos
Envelhecimento/fisiologia , Canais de Cálcio/metabolismo , Cálcio/sangue , Osteoporose/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/urina , Canais de Cálcio/genética , Duodeno/metabolismo , Células Epiteliais/metabolismo , Homeostase/fisiologia , Hiperparatireoidismo/metabolismo , Hiperparatireoidismo/fisiopatologia , Rim/metabolismo , Camundongos , Camundongos Mutantes , Osteocalcina/sangue , Osteoporose/fisiopatologia , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Canais de Cátion TRPV/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA