Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Prostate Cancer Prostatic Dis ; 18(2): 144-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25667107

RESUMO

BACKGROUND: Ketoconazole is a well-known CYP17-targeted systemic treatment for castration-resistant prostate cancer (CRPC). However, most of the published data has been in the pre-chemotherapy setting; its efficacy in the post-chemotherapy setting has not been as widely described. Chemotherapy-naïve patients treated with attenuated doses of ketoconazole (200-300 mg three times daily) had PSA response rate (>50% decline) of 21-62%. We hypothesized that low-dose ketoconazole would likewise possess efficacy and tolerability in the CRPC post-chemotherapy state. METHODS: Men with CRPC and performance status 0-3, adequate organ function and who had received prior docetaxel were treated with low-dose ketoconazole (200 mg orally three times daily) and hydrocortisone (20 mg PO qAM and 10 mg PO qPM) until disease progression. Primary endpoint was PSA response rate (>50% reduction from baseline) where a rate of 25% was to be considered promising for further study (versus a null rate of <5%); 25 patients were required. Secondary endpoints included PSA response >30% from baseline, progression-free survival (PFS), duration of stable disease and evaluation of adverse events (AEs). RESULTS: Thirty patients were accrued with median age of 72 years (range 55-86) and median pre-treatment PSA of 73 ng ml(-1) (range 7-11,420). Twenty-nine patients were evaluable for response and toxicity. PSA response (>50% reduction) was seen in 48% of patients; PSA response (>30% reduction) was seen in 59%. Median PFS was 138 days; median duration of stable disease was 123 days. Twelve patients experienced grade 3 or 4 AEs. Of the 17 grade 3 AEs, only 3 were attributed to treatment. None of the two grade 4 AEs were considered related to treatment. CONCLUSIONS: In docetaxel pre-treated CRPC patients, low-dose ketoconazole and hydrocortisone is a well-tolerated, relatively inexpensive and clinically active treatment option. PSA response to low-dose ketoconazole appears historically comparable to that of abiraterone in this patient context. A prospective, randomized study of available post-chemotherapy options is warranted to assess comparative efficacy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Hidrocortisona/administração & dosagem , Cetoconazol/administração & dosagem , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Taxoides/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Progressão da Doença , Intervalo Livre de Doença , Docetaxel , Humanos , Masculino , Pessoa de Meia-Idade , Antígeno Prostático Específico/sangue , Neoplasias de Próstata Resistentes à Castração/sangue , Neoplasias de Próstata Resistentes à Castração/patologia , Resultado do Tratamento
2.
J Membr Biol ; 212(2): 139-44, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17264983

RESUMO

In normal rats we showed that glucocorticoids participate in the downregulation of UT-A1 protein abundance in the inner medullary tip and in lowering of basal and vasopressin-stimulated facilitated urea permeability in terminal IMCDs. To examine the relevance of this response to a rat model of human disease, we studied rats with uncontrolled diabetes mellitus (DM) induced by streptozotocin (STZ), since these rats have increased corticosterone production and urea excretion. We found that at 3 days of DM, UT-A1 protein abundance is downregulated in the inner medullary tip compared to pair-fed control rats, while DM for more than 7 days caused an increase in UT-A1. To test whether adrenal steroids could be a mechanism contributing to the latter increase, we studied adrenalectomized rats (ADX), ADX rats given STZ to induce diabetes (ADX + STZ), and ADX + STZ rats receiving exogenous aldosterone or dexamethasone. In contrast to control rats, UT-A1 protein abundance was not increased by prolonged DM in the ADX rats. Aquaporin 2 (AQP2) was not increased in the inner medullas of 10-day DM rats either. However, UT-A1 protein abundance was significantly reduced in the inner medullary tips from both diabetic aldosterone-treated (40 +/- 2%) and dexamethasone-treated (43 +/- 2%) ADX rats compared to diabetic ADX rats without steroid replacement. AQP2 was unaffected by steroid hormone treatments. Thus, both mineralocorticoids and glucocorticoids downregulate UT-A1 protein abundance in rats with uncontrolled diabetes mellitus for 10 days. These results suggest that: 1) the increase in UT-A1 observed in DM is dependent upon having adrenal steroids present; and 2) adrenal steroids are not sufficient to enable the compensatory rise in UT-A1 to a steroid-deficient diabetic animal.


Assuntos
Adrenalectomia , Aquaporina 2/metabolismo , Diabetes Mellitus Experimental/metabolismo , Rim/metabolismo , Rim/cirurgia , Proteínas de Membrana Transportadoras/metabolismo , Mineralocorticoides/metabolismo , Adaptação Fisiológica , Animais , Diabetes Mellitus Experimental/induzido quimicamente , Masculino , Ratos , Ratos Sprague-Dawley , Estreptozocina , Regulação para Cima , Transportadores de Ureia
3.
J Anim Sci ; 82(4): 1157-64, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15080338

RESUMO

Urea recycling in ruminants has been studied extensively in the past, but the mechanisms regulating the amount of urea recycled or excreted remain obscure. To elucidate the role of urea transporters (UT) in N recycling, nine Dorset-Finn ewe lambs (20.8 +/- 0.8 kg) were fed diets containing 15.5, 28.4, and 41.3 g of N/kg of DM for 25 d. Nitrogen balance and urea N kinetics were measured during the last 3 d of the period. Animals were then slaughtered and mucosa samples from the rumen, duodenum, ileum, and cecum, as well as kidney medulla and liver, were collected. Increasing N intake tended to increase N balance quadratically (1.5, 5.1, and 4.4 +/- 0.86 g of N/d, P < 0.09), and linearly increased urinary N excretion (2.4, 10, and 16.5 +/- 0.86 g N/d, P < 0.001) and plasma urea N concentration (4.3, 20.3, and 28.4 +/- 2.62 mg of urea N/dL, P < 0.001), but did not affect fecal N excretion (5.0 +/- 0.5 g of N/d; P < 0.94). Urea N production (2.4, 11.8, and 19.2 +/- 0.83 g of N/d; P < 0.001) and urinary urea N excretion (0.7, 7.0, and 13.4 +/- 0.73 g N/d; P < 0.001) increased linearly with N intake, as well as with the urea N recycled to the gastrointestinal tract (1.8, 4.8, and 5.8 +/- 0.40 g of N/d, P < 0.001). No changes due to N intake were observed for creatinine excretion (518 +/- 82.4 mg/d; P < 0.69) and clearance (46 +/- 10.7 mL/min; P < 0.56), but urea N clearance increased linearly with N intake (14.9, 24.4, and 34.9 +/- 5.9 mL/min; P < 0.04). Urea N reabsorption by the kidney tended to decrease (66.3, 38.5, 29.1 +/- 12.6%; P < 0.06) with increasing N content of the diet. Increasing the level of N intake increased linearly the weight of the liver as a proportion of BW (1.73, 1.88, and 2.22 +/- 0.15%, P < 0.03) but only tended to increase the weight of the kidneys (0.36, 0.37, and 0.50 +/- 0.05%, P < 0.08). Urea transporter B was present in all the tissues analyzed, but UT-A was detected only in kidney medulla, liver, and duodenum. Among animals on the three diets, no differences (P > 0.10) in UT abundance, quantified by densitometry, were found. Ruminal-wall urease activity decreased linearly (P < 0.02) with increasing level of N intake. Urease activity in duodenal, ileal, and cecal mucosa did not differ from zero (P > 0.10) in lambs on the high-protein diet. In the present experiment, urea transporter abundance in the kidney medulla and the gastrointestinal tract did not reflect the increase in urea-N reabsorption by the kidney and transferred into the gut.


Assuntos
Rim/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Nitrogênio/administração & dosagem , Nitrogênio/metabolismo , Ovinos/metabolismo , Ureia/metabolismo , Animais , Animais Recém-Nascidos , Nitrogênio da Ureia Sanguínea , Creatinina/urina , Relação Dose-Resposta a Droga , Fezes/química , Feminino , Intestino Delgado/metabolismo , Cinética , Nitrogênio/urina , Tamanho do Órgão , Rúmen/enzimologia , Rúmen/metabolismo , Urease/metabolismo , Transportadores de Ureia
4.
J Membr Biol ; 191(3): 149-63, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12571750

RESUMO

Physiologic data provided evidence for specific urea transporter proteins in red blood cells and kidney inner medulla. During the past decade, molecular approaches resulted in the cloning of several urea transporter cDNA isoforms derived from two gene families: UT-A and UT-B. Polyclonal antibodies were generated to the cloned urea transporter proteins, and their use in integrative animal studies resulted in several novel findings, including: (1) UT-B is the Kidd blood group antigen; (2) UT-B is also expressed in many non-renal tissues and endothelial cells; (3) vasopressin increases UT-A1 phosphorylation in rat inner medullary collecting duct; (4) the surprising finding that UT-A1 protein abundance and urea transport are increased in the inner medulla during conditions in which urine concentrating ability is reduced; and (5) UT-A protein abundance is increased in uremia in both liver and heart. This review will summarize the knowledge gained from studying molecular mechanisms of urea transport and from integrative studies into urea transporter protein regulation.


Assuntos
Homeostase/fisiologia , Medula Renal/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Ureia/farmacocinética , Angiotensinas/farmacologia , Animais , Transporte Biológico Ativo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Diabetes Mellitus/fisiopatologia , Humanos , Rim/efeitos dos fármacos , Rim/fisiologia , Capacidade de Concentração Renal/efeitos dos fármacos , Capacidade de Concentração Renal/fisiologia , Medula Renal/efeitos dos fármacos , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/classificação , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Ratos , Insuficiência Renal/fisiopatologia , Ureia/química , Ureia/metabolismo , Uremia/fisiopatologia , Urina , Vasopressinas/farmacologia , Transportadores de Ureia
5.
Kidney Int ; 60(6): 2274-82, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11737600

RESUMO

BACKGROUND: Cisplatin (CP) induced polyuria in rats is associated with a reduction in medullary hypertonicity, normally generated by the thick ascending limb (TAL) salt transporters, and the collecting duct urea transporters (UT). To investigate the molecular basis of this abnormality, we determined the protein abundance of major salt and UT isoforms in rat kidney during CP-induced polyuria. METHODS: Male Sprague-Dawley rats received either a single injection of CP (5 mg/kg, N = 6) or saline (N = 6) intraperitoneally five days before sacrifice. Urine, blood, and kidneys were collected and analyzed. RESULTS: CP-treated rats developed polyuric acute renal failure as assessed by increased blood urea nitrogen (BUN), urine volume and decreased urine osmolality. Western analysis of kidney homogenates revealed a marked reduction in band density of the bumetanide-sensitive Na-K-2Cl cotransporter in cortex (60% of control values, P < 0.05), but not in outer medulla (OM) (106% of control values). There were no differences in band densities for the renal outer medullary potassium channel (ROMK), the type III Na-H exchanger (NHE3), the alpha-subunit of Na,K-ATPase in the OM; or for UT-A1, UT-A2 or UT-A4 in outer or inner medulla. However, the band pattern of UT-A2 and UT-A4 proteins in the OM of CP-treated rats was different from the control rats, suggesting a qualitative modification of these proteins. CONCLUSIONS: Changes in the abundance of outer or inner medullary salt or urea transporters are unlikely to play a role in the CP-induced reduction in medullary hypertonicity. However, qualitative changes in UT proteins may affect their functionality and thus may have a role.


Assuntos
Antineoplásicos , Proteínas de Transporte/metabolismo , Cisplatino , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Poliúria/induzido quimicamente , Poliúria/metabolismo , Cloreto de Sódio/metabolismo , Animais , Nitrogênio da Ureia Sanguínea , Northern Blotting , Peso Corporal , Immunoblotting , Rim/patologia , Masculino , Platina/farmacocinética , Poliúria/patologia , Ratos , Ratos Sprague-Dawley , Urina/química , Transportadores de Ureia
6.
Am J Physiol Cell Physiol ; 281(4): C1318-25, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11546670

RESUMO

A new polyclonal antibody to the human erythrocyte urea transporter UT-B detects a broad band between 45 and 65 kDa in human erythrocytes and between 37 and 51 kDa in rat erythrocytes. In human erythrocytes, the UT-B protein is the Kidd (Jk) antigen, and Jk(a+b+) erythrocytes express the 45- to 65-kDa band. However, in Jk null erythrocytes [Jk(a-b-)], only a faint band at 55 kDa is detected. In kidney medulla, a broad band between 41 and 54 kDa, as well as a larger band at 98 kDa, is detected. Human and rat kidney show UT-B staining in nonfenestrated endothelial cells in descending vasa recta. UT-B protein and mRNA are detected in rat brain, colon, heart, liver, lung, and testis. When kidney medulla or liver proteins are analyzed with the use of a native gel, only a single protein band is detected. UT-B protein is detected in cultured bovine endothelial cells. We conclude that UT-B protein is expressed in more rat tissues than previously reported, as well as in erythrocytes.


Assuntos
Proteínas de Transporte/análise , Proteínas de Transporte/imunologia , Eritrócitos/química , Rim/química , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana Transportadoras , Sequência de Aminoácidos , Animais , Anticorpos , Aorta/química , Química Encefálica , Proteínas de Transporte/genética , Colo/química , Eletroforese em Gel de Poliacrilamida , Endotélio Vascular/química , Expressão Gênica/fisiologia , Humanos , Pulmão/química , Masculino , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Miocárdio/química , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Organismos Livres de Patógenos Específicos , Testículo/química , Transportadores de Ureia
7.
Am J Physiol Renal Physiol ; 281(3): F400-6, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11502588

RESUMO

We have isolated and characterized the human homolog of the rat largest urea transporter of the UT-A family (hUT-A1). The 4.2-kb hUT-A1 cDNA encodes a 920-amino acid peptide, which is 89% identical to the rat UT-A1 protein. By Northern hybridization, hUT-A1 expression is detected in the human inner medulla as a approximately 4.4-kb mRNA transcript. By Western analysis, hUT-A1 is identified as a approximately 100-kDa protein in the human inner medulla. By immunohistochemistry, hUT-A1 expression is localized to the inner medullary collecting duct (IMCD). When transfected into HEK-293 cells hUT-A1 cDNA is translated into a approximately 98-kDa protein. Expression of hUT-A1 in Xenopus oocytes results in phloretin-inhibitable uptake of (14)C-urea, which shows only modest stimulation by cAMP, suggesting that in the human IMCD vasopressin may have a limited role in the short-term regulation of hUT-A1-mediated urea transport. We determined the organization of the human Slc14a2 gene and identified 20 exons distributed over approximately 67.5 kb on chromosome 18, from which hUT-A1 and the other human urea transporter, hUT-A2, are transcribed.


Assuntos
Proteínas de Transporte/genética , Cromossomos Humanos Par 18 , Medula Renal/fisiologia , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras , Sequência de Aminoácidos , Animais , Proteínas de Transporte/análise , Proteínas de Transporte/química , Linhagem Celular , Mapeamento Cromossômico , Clonagem Molecular , Éxons , Humanos , Túbulos Renais Coletores/fisiologia , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/química , Dados de Sequência Molecular , Biossíntese de Proteínas , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Transfecção , Ureia/metabolismo , Transportadores de Ureia
8.
Circ Res ; 89(2): 139-45, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11463720

RESUMO

Urea transporters have been cloned from kidney medulla (UT-A) and erythrocytes (UT-B). We determined whether UT-A proteins could be detected in heart and whether their abundance was altered by uremia or hypertension or in human heart failure. In normal rat heart, bands were detected at 56, 51, and 39 kDa. In uremic rats, the abundance of the 56-kDa protein increased 1.9-fold compared with pair-fed, sham-operated rats, whereas the 51- and 39-kDa proteins were unchanged. We also detected UT-A2 mRNA in hearts from control and uremic rats. Because uremia is accompanied by hypertension, the effects of hypertension per se were studied in uninephrectomized deoxycorticosterone acetate salt-treated rats, where the abundance of the 56-kDa protein increased 2-fold versus controls, and in angiotensin II-infused rats, where the abundance of the 56 kDa protein increased 1.8-fold versus controls. The 51- and 39-kDa proteins were unchanged in both hypertensive models. In human left ventricle myocardium, UT-A proteins were detected at 97, 56, and 51 kDa. In failing left ventricle (taken at transplant, New York Heart Association class IV), the abundance of the 56-kDa protein increased 1.4-fold, and the 51-kDa protein increased 4.3-fold versus nonfailing left ventricle (donor hearts). We conclude that (1) multiple UT-A proteins are detected in rat and human heart; (2) the 56-kDa protein is upregulated in rat heart in uremia or models of hypertension; and (3) the rat results can be extended to human heart, where 56- and 51-kDa proteins are increased during heart failure.


Assuntos
Proteínas de Transporte/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Miocárdio/metabolismo , Adulto , Animais , Western Blotting , Proteínas de Transporte/genética , Feminino , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Humanos , Hipertensão/genética , Hipertensão/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Pessoa de Meia-Idade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Uremia/genética , Uremia/metabolismo , Transportadores de Ureia
9.
Am J Physiol Renal Physiol ; 281(1): F133-43, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11399654

RESUMO

UT-A1 is an extremely hydrophobic 929-amino acid integral membrane protein, expressed in the renal inner medullary collecting duct, with a central role in the urinary concentrating mechanism. Previous immunoblotting studies in rats have revealed that UT-A1 is present in kidney in 97- and 117-kDa monomeric forms and that the relative abundance of the two forms is altered by vasopressin treatment and other treatments that altered urinary inner medullary urea concentration. The present studies were carried out using protein chemistry techniques to determine the origin of the two forms. Peptide-directed polyclonal antibodies targeted to five sites along the polypeptide sequence from the NH2 to the COOH terminus labeled both forms, thus failing to demonstrate a significant deletion in the primary amino acid chain. The 97- and 117-kDa monomeric forms were both reduced to 88 kDa by deglycosylation with N-glycosidase F, indicating that a single polypeptide chain is glycosylated to two different extents. Studies using nonionic detergents for membrane solubilization or using homobifunctional cross-linkers demonstrated that UT-A1 exists as a 206-kDa protein complex in native kidney membranes. The mobility of this complex was also increased by deglycosylation. Both the 97- and 117-kDa proteins, as well as the 206-kDa complex, were immunoprecipitated with UT-A1 antibodies. We conclude that UT-A1 is a glycoprotein and that the two monomeric forms (97 and 117 kDa) in inner medullary collecting duct are the consequence of different states of glycosylation.


Assuntos
Proteínas de Transporte/análise , Túbulos Renais Coletores/metabolismo , Glicoproteínas de Membrana/análise , Proteínas de Membrana Transportadoras , Animais , Anticorpos/imunologia , Sítios de Ligação , Proteínas de Transporte/química , Proteínas de Transporte/imunologia , Membrana Celular/metabolismo , Reagentes de Ligações Cruzadas , Eletroforese , Epitopos/imunologia , Glicosilação , Hexosaminidases/farmacologia , Immunoblotting , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/imunologia , Testes de Precipitina , Isoformas de Proteínas/química , Ratos , Ratos Sprague-Dawley , Vasopressinas/farmacologia , Transportadores de Ureia
10.
Biochim Biophys Acta ; 1518(1-2): 19-26, 2001 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-11267655

RESUMO

We cloned the Slc14a2 gene and determined the genomic organization of the rat urea transporter UT-A. Slc14a2, the gene encoding the rat UT-A transporter, extends for more that 300 kb. The four known rat mRNA isoforms: UT-A1, UT-A2, UT-A3, and UT-A4 are transcribed from 24 exons. The Slc14a2 genomic map also accounts for 3'-untranslated sequences expressed alternatively in UT-A1, UT-A2, and UT-A3. We previously identified a TATA-less, tonicity-responsive promoter controlling the transcription of UT-A1, UT-A3, and UT-A4 from a single initiation site in the 5'-flanking region of the gene. Here, we describe a second, internal promoter in intron 12, which controls the transcription of UT-A2 starting from exon 13. This region contains a TATA motif upstream from the UT-A2 transcription start site, and shows consensus sequences for the cAMP response element (CRE) and for the tonicity enhancer (TonE) motif. Stimulation by cAMP induces UT-A2 mRNA expression in mIMCD3 cells, and luciferase activity in mIMCD3 cells transfected with those pGL3 constructs including the CRE sequences. Although long-term exposure to hypertonicity induces UT-A2 expression in mIMCD3 cells, hypertonicity does not induce significantly the activity of the promoter in intron 12. In summary, we describe the genomic structure of the rat UT-A urea transporter, encoded by the Slc14a2 gene. Our findings suggest that two promoters regulate transcription of the four UT-A isoforms, and that stimulation of transcription by vasopressin, mediated by cAMP and CRE sequences, and controlled by an intronic promoter, may contribute to the increase in UT-A2 expression during water deprivation.


Assuntos
Proteínas de Transporte/genética , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras , Ureia/metabolismo , Animais , Sequência de Bases , Proteínas de Transporte/metabolismo , Clonagem Molecular , AMP Cíclico/metabolismo , Glicoproteínas de Membrana/metabolismo , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ratos , Transcrição Gênica , Transportadores de Ureia
11.
Am J Physiol Renal Physiol ; 279(5): F835-40, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11053043

RESUMO

Angiotensin II receptors are present along the rat inner medullary collecting duct (IMCD), although their physiological role is unknown. Because urea is one of the major solutes transported across the terminal IMCD, we measured angiotensin II's effect on urea permeability. In the perfused rat terminal IMCD, angiotensin II had no effect on basal urea permeability but significantly increased vasopressin-stimulated urea permeability by 55%. Angiotensin II, both without and with vasopressin, also increased the amount of (32)P incorporated into urea transporter (UT)-A1 in inner medullary tissue exposed to these hormones ex vivo. Because angiotensin II activates protein kinase C, we tested the effect of staurosporine (SSP). In the absence of angiotensin II, SSP had no effect on vasopressin-stimulated urea permeability in the perfused terminal IMCD. However, SSP completely and reversibly blocked the angiotensin II-mediated increase in vasopressin-stimulated urea permeability. SSP and chelerythrine reduced the angiotensin II-stimulated (32)P incorporation into UT-A1 in inner medullary tissue exposed ex vivo. We conclude that angiotensin II increases vasopressin-stimulated facilitated urea permeability and (32)P incorporation into the 97- and 117-kDa UT-A1 proteins via a protein kinase C-mediated signaling pathway. These data suggest that angiotensin II augments vasopressin-stimulated facilitated urea transport in the rat terminal IMCD and may play a physiological role in the urinary concentrating mechanism by augmenting the maximal response to vasopressin.


Assuntos
Angiotensina II/metabolismo , Medula Renal/metabolismo , Túbulos Renais Coletores/metabolismo , Proteínas de Membrana Transportadoras , Ureia/metabolismo , Vasopressinas/metabolismo , Angiotensina II/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Proteínas de Transporte/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Glicoproteínas de Membrana/metabolismo , Concentração Osmolar , Permeabilidade/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Vasopressinas/farmacologia , Transportadores de Ureia
12.
J Biol Chem ; 275(49): 38275-80, 2000 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-10995747

RESUMO

The rat renal urea transporter UT-A includes four isoforms. UT-A1, UT-A3, and UT-A4 are transcribed from a single initiation site at the 5'-end of the gene; a distinct internal initiation site is used for UT-A2 transcription. We cloned 1.3 kilobases (kb) of the 5'-flanking region upstream of the transcription start site of UT-A1, UT-A3, and UT-A4. This region contains three CCAAT sequences but lacks a TATA motif. A tonicity-responsive enhancer (TonE) was identified at -377bp. The 1.3-kb full fragment subcloned into pGL3 vector induced luciferase activity in Madin-Darby canine kidney cells and in mouse inner medullary collecting duct cells in isotonic medium. Luciferase activity was increased significantly in hypertonic medium, whereas deletion or mutation of the TonE sequence abolished this response. Electrophoretic mobility shift assay using the 5' UT-A TonE sequence as DNA probe showed formation of a specific DNA-protein complex with nuclear extracts from cells exposed to hypertonic medium and was weakly detectable in isotonic controls. A supershift in the mobility of the DNA-protein complex was observed with antiserum targeted to the TonE-binding protein (TonEBP). Co-transfection with dominant-negative TonEBP abolished the luciferase activity induced by the UT-A 1.3-kb construct under hypertonic and isotonic conditions. These data suggest that the TonE/TonEBP pathway mediates tonicity-responsive transcriptional regulation of UT-A1, UT-A3, and UT-A4 expression.


Assuntos
Proteínas de Transporte/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras , Transativadores/metabolismo , Transcrição Gênica , Animais , Sequência de Bases , Linhagem Celular , Clonagem Molecular , Cães , Medula Renal/metabolismo , Túbulos Renais Coletores/metabolismo , Luciferases/genética , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Isoformas de Proteínas/genética , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Deleção de Sequência , Fatores de Transcrição , Transfecção , Transportadores de Ureia
13.
Mt Sinai J Med ; 67(2): 112-9, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10747366

RESUMO

Due to urea's role in producing concentrated urine, its transport is critically important to the conservation of body water. Within the renal inner medulla, urea is transported by both facilitated and active urea transport mechanisms. The vasopressin-regulated, facilitated urea transporter (UT-A1) in the terminal inner medullary collecting duct (IMCD) permits high rates of transepithelial urea transport and results in delivery of large quantities of urea into the deepest portions of the inner medulla where it is needed to maintain a high interstitial osmolality for maximal urine concentration. Four cDNA isoforms of the UT-A urea transporter family have been cloned. In addition, there are three secondary active, sodium-dependent, urea transport mechanisms in IMCD subsegments: (1) active urea secretion in the apical membrane of the terminal IMCD from untreated rats; (2) active urea absorption in the apical membrane of the initial IMCD from low-protein fed or hypercalcemic rats; and (3) active urea absorption in the basolateral membrane of the initial IMCD from furosemide-treated rats. This review will focus on integrative studies of the rapid and long-term regulation of urea transporters in rats with reduced urine concentrating ability. These studies led to the surprising result that the basal-facilitated urea permeability in the terminal IMCD and UT-A1 protein abundance are increased during in vivo conditions associated with an impaired urine concentrating ability. In contrast, there are two response patterns of active urea transporters: (1) hypercalcemia, a low-protein diet, and furosemide result in induction of active urea absorption in the initial IMCD, albeit by different mechanisms, and inhibition of active urea secretion in the terminal IMCD; while (2) water diuresis results in up-regulation of active urea secretion in the terminal IMCD without any active urea absorption in the initial IMCD. The first pattern contributes to the urine concentrating defect by increasing urea delivery to the base of the inner medulla, thus decreasing urea delivery distally to the inner medullary tip. The second response pattern will directly decrease urea content in the deep inner medulla. UT-A urea transporters are also expressed outside the kidney. Recent studies show that the liver has phloretin-inhibitable urea transport and that it occurs via a 49 kDa UT-A protein. When rats are made uremic, the abundance of this 49 kDa UT-A protein increases in the liver in vivo. This up-regulation of the 49 kDa UT-A protein may allow hepatocytes to increase ureagenesis to reduce the accumulation of ammonium and/or bicarbonate in uremia.


Assuntos
Proteínas de Transporte/fisiologia , Capacidade de Concentração Renal/fisiologia , Rim/fisiologia , Fígado/fisiologia , Ureia/metabolismo , Animais , Transporte Biológico Ativo , Diabetes Mellitus Experimental/metabolismo , RNA Mensageiro/metabolismo , Ratos , Vasopressinas/fisiologia
14.
J Gerontol A Biol Sci Med Sci ; 55(1): B26-34, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10719760

RESUMO

To determine if the aging-associated decline in testosterone results in attenuated vasopressin (VP) responses to dehydration, testosterone implants were given to aged male Fischer 344Brown-Norway F1(F344BNF1) rats. Water deprivation caused comparable dehydration, increased plasma VP (pVP), and decreased posterior pituitary (PP) VP content in 4-, 15-, and 28-month-old rats. Dehydration increased VP mRNA content of supraoptic nuclei only at 4 months, whereas VP mRNA length was increased at both 4 and 15 months of age. The elevated pVP in the water-deprived aged rats indicates that even without an increase in VP mRNA content, PP VP storage was adequate to maintain elevated pVP. Dehydration increased aquaporin-2 content at 4, but not at 15 or 28 months of age, suggesting decreased renal responsiveness to VP. Testosterone replacement did not produce dehydration-induced increases in VP mRNA or aquaporin-2. Therefore, testosterone deficiency does not result in altered VP responses to dehydration in aged F344BNF1 rats.


Assuntos
Envelhecimento/fisiologia , Aquaporinas/metabolismo , Desidratação/fisiopatologia , Testosterona/farmacologia , Vasopressinas/metabolismo , Análise de Variância , Animais , Northern Blotting , Western Blotting , Desidratação/metabolismo , Implantes de Medicamento , Medula Renal/metabolismo , Masculino , Neuro-Hipófise/metabolismo , Progesterona/sangue , Progesterona/metabolismo , RNA/análise , Radioimunoensaio , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344 , Vasopressinas/sangue
15.
J Am Soc Nephrol ; 10(10): 2076-83, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10505683

RESUMO

In perfused rat liver, there is phloretin-inhibitable urea efflux, but whether it is mediated by the kidney UT-A urea transporter family is unknown. To determine whether cultured HepG2 cells transport urea, thiourea influx was measured. HepG2 cells had a thiourea influx rate of 1739 +/- 156 nmol/g protein per min; influx was inhibited 46% by phloretin and 32% by thionicotinamide. Western analysis of HepG2 cell lysate using an antibody to UT-A1, UT-A2, and UT-A4 revealed two protein bands: 49 and 36 kD. The same bands were detected in cultured rat hepatocytes, freshly isolated rat hepatocytes, and in liver from rat, mouse, and chimpanzee. Both bands were present when analyzed by native gel electrophoresis, and deglycosylation of rat liver lysate had no effect on either band. Differential centrifugation of rat liver lysate showed that the 49-kD protein is in the membrane fraction and the 36-kD protein is in the cytoplasm. To determine whether the abundance of these UT-A proteins varies in vivo, rats were made uremic by 5/6 nephrectomy. The 49-kD protein was significantly increased 5.5-fold in livers from uremic rats compared to pair-fed control rats. It is concluded that phloretin-inhibitable urea flux in liver may occur via a 49-kD protein that is specifically detected by a UT-A antibody. Uremia increases the abundance of this 49-kD UT-A protein in rat liver in vivo.


Assuntos
Proteínas de Transporte/metabolismo , Fígado/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Uremia/metabolismo , Animais , Transporte Biológico , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Fígado/citologia , Masculino , Camundongos , Nefrectomia , Ratos , Ratos Sprague-Dawley , Valores de Referência , Sensibilidade e Especificidade , Tioureia/análise , Regulação para Cima , Transportadores de Ureia
16.
Curr Opin Nephrol Hypertens ; 8(4): 499-504, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10491746

RESUMO

The terminal inner medullary collecting duct facilitated urea transporter, UT-A1, belongs to a family of four urea transporters. The inner medullary collecting duct also expresses three active urea transporters. Reducing urine concentrating ability in vivo upregulates UT-A1 and alters active urea secretion in the terminal inner medullary collecting duct, and induces active urea reabsorption in the initial inner medullary collecting duct.


Assuntos
Proteínas de Transporte/fisiologia , Medula Renal/fisiologia , Túbulos Renais Coletores/fisiologia , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana Transportadoras , Ureia/metabolismo , Animais , Proteínas de Transporte/genética , Diabetes Mellitus Experimental/fisiopatologia , Regulação da Expressão Gênica , Humanos , Glicoproteínas de Membrana/genética , Síndrome Nefrótica/fisiopatologia , Urina/fisiologia , Transportadores de Ureia
18.
J Am Soc Nephrol ; 10(2): 230-7, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10215321

RESUMO

Urea transport in the kidney is important for the production of concentrated urine and is mediated by a family of transporter proteins, identified from erythropoietic tissue (UT-B) and from kidney (UT-A). Two isoforms of the renal urea transporter (UT-A) have been cloned so far: UT-A1 and UT-A2. We used rapid amplification of cDNA ends to clone two new isoforms of the rat UT-A transporter: UT-A3 and UT-A4. UT-A3 and UT-A4 are 87% homologous. The UT-A3 cDNA encodes a peptide of 460 amino acids, which corresponds to the amino-terminal half of the UT-A1 peptide and is 62% identical to UT-A2. The UT-A4 cDNA encodes a peptide of 466 amino acids, which is 84% identical to UT-A2. Transient transfection of HEK-293 cells with the UT-A3 or UT-A4 cDNA results in phloretin-inhibitable urea uptake, which is increased by forskolin. Thus, both new isoforms encode functional urea transporters that may be vasopressin-regulated. UT-A3 and UT-A4 mRNA are expressed in the renal outer and inner medulla but not in the cortex; unidentified UT-A isoforms similar to UT-A3 may also be expressed in the testis. It is concluded that there are at least four different rat UT-A urea transporters.


Assuntos
Proteínas de Transporte/genética , Clonagem Molecular , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras , Ratos/genética , Sequência de Aminoácidos/genética , Animais , Sequência de Bases/genética , Linhagem Celular , DNA Complementar/genética , Humanos , Rim/citologia , Rim/embriologia , Rim/metabolismo , Dados de Sequência Molecular , Biossíntese de Proteínas/genética , RNA Mensageiro/metabolismo , Distribuição Tecidual , Transportadores de Ureia
20.
J Am Soc Nephrol ; 10(3): 635-46, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10073615

RESUMO

Urea is important for the conservation of body water due to its role in the production of concentrated urine in the renal inner medulla. Physiologic data demonstrate that urea is transported by facilitated and by active urea transporter proteins. The facilitated urea transporter (UT-A) in the terminal inner medullary collecting duct (IMCD) permits very high rates of transepithelial urea transport and results in the delivery of large amounts of urea into the deepest portions of the inner medulla where it is needed to maintain a high interstitial osmolality for concentrating the urine maximally. Four isoforms of the UT-A urea transporter family have been cloned to date. The facilitated urea transporter (UT-B) in erythrocytes permits these cells to lose urea rapidly as they traverse the ascending vasa recta, thereby preventing loss of urea from the medulla and decreasing urine-concentrating ability by decreasing the efficiency of countercurrent exchange, as occurs in Jk null individuals (who lack Kidd antigen). In addition to these facilitated urea transporters, three sodium-dependent, secondary active urea transport mechanisms have been characterized functionally in IMCD subsegments: (1) active urea reabsorption in the apical membrane of initial IMCD from low-protein fed or hypercalcemic rats; (2) active urea reabsorption in the basolateral membrane of initial IMCD from furosemide-treated rats; and (3) active urea secretion in the apical membrane of terminal IMCD from untreated rats. This review focuses on the physiologic, biophysical, and molecular evidence for facilitated and active urea transporters, and integrative studies of their acute and long-term regulation in rats with reduced urine-concentrating ability.


Assuntos
Água Corporal/metabolismo , Proteínas de Transporte/metabolismo , Capacidade de Concentração Renal/fisiologia , Túbulos Renais Coletores/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Ureia/metabolismo , Animais , Proteínas de Transporte/farmacologia , Humanos , Capacidade de Concentração Renal/efeitos dos fármacos , Testes de Função Renal , Glicoproteínas de Membrana/farmacologia , Concentração Osmolar , Coelhos , Ratos , Equilíbrio Hidroeletrolítico , Transportadores de Ureia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA