Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
J Frailty Aging ; 11(4): 348-369, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36346721

RESUMO

OBJECTIVES: To present the local evidence and final recommendations of the Clinical Practice Guidelines workgroup convened by the Chapter of Geriatricians and the Society for Geriatric Medicine Singapore. The aim is to develop contextualized evidence-based recommendations that facilitate adoption of the Asian Working Group for Sarcopenia (AWGS) 2019 consensus into current practice in Singapore. METHODS: The workgroup drew upon the AWGS'2019 consensus, updated literature review of Singapore studies till 31 Dec 2020, and evidence from recent systematic reviews. From 40 local studies included for data extraction, we constructed evidence tables organized as: definition and epidemiology; diagnosis and evaluation; and treatment and intervention. Twenty recommendations - case-finding, diagnosis, treatment, prevention, research - were developed, and graded for strength and quality using the GRADE approach. Consensus from an expert panel(N=23) was achieved after two rounds of the modified Delphi process. RESULTS: The local prevalence of sarcopenia among community-dwelling older adults ranged from 13.6% to 25%. Most studies adopted the AWGS'2019 and AWGS'2014 criteria. Reported case finding tools include SARC-F, calf circumference (CC) and SARC-CalF. Gender-specific AWGS cut-offs for appendicular skeletal mass were used to define low muscle mass. Different protocols and dynamometers were used to assess handgrip strength, whilst gait speed and 5-times chair stand were commonly used to assess physical performance. RECOMMENDATIONS: We conditionally recommend a case-finding approach in at-risk older adults using validated case-finding tools. Screen-positive individuals should be assessed for 'possible sarcopenia' and underlying causes. For diagnosis, we conditionally recommend using the AWGS'2019 algorithm, and dual-energy X-ray absorptiometry when necessary to determine low lean mass for a confirmatory diagnosis of sarcopenia. For treatment, we strongly recommend resistance-based exercises and conditionally recommend a quality protein-rich diet/protein supplementation, with Vitamin D supplementation for insufficiency (<30 micrograms/L). For prevention, we recommend regular resistance-based physical activity and adequate protein intake (≥1.0g/kg bodyweight). We encourage more research to address local evidence gaps.


Assuntos
Sarcopenia , Humanos , Idoso , Sarcopenia/diagnóstico , Sarcopenia/epidemiologia , Sarcopenia/prevenção & controle , Força da Mão , Singapura/epidemiologia , Força Muscular/fisiologia , Velocidade de Caminhada , Programas de Rastreamento/métodos , Avaliação Geriátrica/métodos
2.
Am J Physiol Regul Integr Comp Physiol ; 288(4): R1046-56, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15563580

RESUMO

Selective transporters account for rapid urea transport across plasma membranes of several cell types. UT-B1 urea transporter is widely distributed in rat and human tissues. Because mice exhibit high urea turnover and are the preferred species for gene engineering, we have delineated UT-B1 tissue expression in murine tissues. A cDNA was cloned from BALB/c mouse kidney, encoding a polypeptide that differed from C57BL/6 mouse UT-B1 by one residue (Val-8-Ala). UT-B1 mRNA was detected by RT-PCR in brain, kidney, bladder, testis, lung, spleen, and digestive tract (liver, stomach, jejunum, colon). Northern blotting revealed seven UT-B1 transcripts in mouse tissues. Immunoblots identified a nonglycosylated UT-B1 protein of 29 kDa in most tissues and of 36 and 32 kDa in testis and liver, respectively. UT-B1 protein of gastrointestinal tract did not undergo N-glycosylation. Immunohistochemistry and in situ hybridization localized UT-B1 in urinary tract urothelium (papillary surface, ureter, bladder, and urethra), prominently on plasma membranes and restricted to the basolateral area in umbrella cells. UT-B1 was found in endothelial cells of descending vasa recta in kidney medulla and in astrocyte processes in brain. Dehydration induced by water deprivation for 2 days caused a tissue-specific decrease in UT-B1 abundance in the urinary bladder and the ureter.


Assuntos
Sistema Digestório/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Sistema Urinário/metabolismo , Animais , Northern Blotting , Western Blotting , Clonagem Molecular , DNA Complementar/biossíntese , DNA Complementar/genética , Desidratação/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Rim/metabolismo , Masculino , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sede/fisiologia , Ureter/metabolismo , Bexiga Urinária/metabolismo , Urotélio/metabolismo
3.
Am J Physiol Regul Integr Comp Physiol ; 285(6): R1355-65, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12933359

RESUMO

Aging is commonly associated with defective urine-concentrating ability. The present study examined how the kidney and the brain of senescent (30-mo-old) female WAG/Rij rats respond to dehydration induced by 2 days of water deprivation in terms of urea transporter (UT) regulation. In euhydrated situation, senescent rats exhibited similar vasopressin plasma level but lower urine osmolality and papillary urea concentration and markedly reduced kidney UT-A1, UT-A3, and UT-B1 abundances compared with adult (10-mo-old) rats. Senescent rats responded to dehydration similarly to adult rats by a sixfold increase in vasopressin plasma level. Their papillary urea concentration was doubled, without, however, attaining that of dehydrated adult rats. Such an enhanced papillary urea sequestration occurred with a great fall of both UT-A1 and UT-A3 abundances in the tip of inner medulla and an increased UT-A1 abundance in the base of inner medulla. UT-A2 and UT-B1 were unchanged. These data suggest that the inability of control and thirsted senescent rats to concentrate urine as much as their younger counterparts derives from lower papillary urea concentration. In aging brain, UT-B1 abundance was increased twofold together with a fourfold increase in aquaporin-4 abundance. Dehydration did not alter the abundance of these transporters.


Assuntos
Envelhecimento/fisiologia , Desidratação/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Ureia/metabolismo , Animais , Aquaporina 4 , Aquaporinas/metabolismo , Arginina Vasopressina/sangue , Encéfalo/metabolismo , Feminino , Capacidade de Concentração Renal/fisiologia , Medula Renal/metabolismo , Concentração Osmolar , Ratos , Ratos Endogâmicos , Privação de Água/fisiologia , Transportadores de Ureia
4.
Am J Physiol Renal Physiol ; 283(5): F912-22, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12372766

RESUMO

UT-B1 is the facilitated urea transporter of red blood cells (RBCs) and endothelial cells of descending vasa recta in the kidney. Immunoblotting with a polyclonal antibody against the C-ter sequence of rat UT-B1 revealed UT-B1 as both nonglycosylated (29 kDa) and N-glycosylated (47.5 and 33 kDa) proteins in RBC membranes, kidney medulla, brain, and bladder in rat. In testis, UT-B1 was expressed only as a nonglycosylated protein of 47.5 kDa. Immunocytochemistry confirmed that the location of UT-B1 is restricted to descending vasa recta. In brain, UT-B1 protein was found in astrocytes and ependymal cells. Cell bodies and perivascular end feet of astrocytes were labeled in brain cortex, whereas astrocyte cell processes were labeled in corpus callosum. Flow cytometry analysis of RBCs revealed a good cross-reactivity of the antibody with mouse and human UT-B1. UT-B1 protein expression in rat kidney medulla was downregulated greatly by long-term [deamino-Cys(1),D-Arg(8)]vasopressin infusion and moderately by furosemide treatment. This study discloses an uneven distribution of UT-B1 protein within astrocytes and the regulation of renal UT-B1 protein by antidiuretic hormone.


Assuntos
Proteínas de Transporte/metabolismo , Eritrócitos/metabolismo , Rim/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Vasopressinas/metabolismo , Sequência de Aminoácidos , Animais , Astrócitos/metabolismo , Proteínas de Transporte/análise , Proteínas de Transporte/imunologia , Desamino Arginina Vasopressina/farmacologia , Diuréticos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Eritrócitos/química , Furosemida/farmacologia , Glicosilação , Humanos , Imuno-Histoquímica , Rim/química , Medula Renal/química , Medula Renal/metabolismo , Masculino , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Coelhos , Ratos , Ratos Sprague-Dawley , Fármacos Renais/farmacologia , Especificidade da Espécie , Transportadores de Ureia
5.
Am J Physiol Renal Physiol ; 281(6): F1123-31, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11704564

RESUMO

The mechanisms underlying the prevention of age-related polyuria by chronic food restriction were investigated in female WAG/Rij rats. The decreased osmolality of renal papilla observed in senescent rats was not corrected by food restriction. A reduced urea content in the inner medulla of senescent rats, fed ad libitum or food-restricted, was suggested by the marked decrease in expression of UT-A1 and UT-B1 urea transporters. Aquaporin-2 (AQP2) downregulation in the inner medulla of senescent rats was partially prevented by food restriction. Both AQP2 and the phosphorylated form of AQP2 (p-AQP2), the presence of which was diffuse within the cytoplasm of collecting duct principal cells in normally fed senescent rats, were preferentially targeted at the apical region of the cells in food-restricted senescent animals. Plasma vasopressin (AVP) was similar in 10- and 30-mo-old rats fed ad libitum, but was doubled in food-restricted 30-mo-old rats. This study indicates that 1) kidney aging is associated with a marked decrease in AQP2, UT-A1, and UT-B1 expression in the inner medulla and a reduced papillary osmolality; and 2) the prevention of age-related polyuria by chronic food restriction occurs through an improved recruitment of AQP2 and p-AQP2 to the apical membrane in inner medulla principal cells, permitted by increased plasma AVP concentration.


Assuntos
Envelhecimento , Aquaporinas/metabolismo , Ingestão de Alimentos , Rim/metabolismo , Proteínas de Membrana Transportadoras , Poliúria/prevenção & controle , Vasopressinas/fisiologia , Animais , Aquaporina 2 , Aquaporina 6 , Proteínas de Transporte/metabolismo , Polaridade Celular , Feminino , Medula Renal/química , Medula Renal/metabolismo , Medula Renal/ultraestrutura , Túbulos Renais/metabolismo , Túbulos Renais/ultraestrutura , Glicoproteínas de Membrana/metabolismo , Concentração Osmolar , Fosforilação , Poliúria/etiologia , Poliúria/metabolismo , Transporte Proteico , Ratos , Urina/química , Vasopressinas/sangue , Desequilíbrio Hidroeletrolítico/etiologia , Desequilíbrio Hidroeletrolítico/metabolismo , Transportadores de Ureia
6.
Leukemia ; 15(9): 1398-407, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11516100

RESUMO

Lovastatin is an inhibitor of the enzyme 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, the major regulatory enzyme of the mevalonate pathway. We have previously reported that lovastatin induces a significant apoptotic response in human acute myeloid leukemia (AML) cells. To identify the critical biochemical mechanism(s) essential for lovastatin-induced apoptosis, add-back experiments were conducted to determine which downstream product(s) of the mevalonate pathway could suppress this apoptotic response. Apoptosis induced by lovastatin was abrogated by mevalonate (MVA) and geranylgeranyl pyrophosphate (GGPP), and was partially inhibited by farnesyl pyrophosphate (FPP). Other products of the mevalonate pathway including cholesterol, squalene, lanosterol, desmosterol, dolichol, dolichol phosphate, ubiquinone, and isopentenyladenine did not affect lovastatin-induced apoptosis in AML cells. Our results suggest that inhibiting geranylgeranylation of target proteins is the predominant mechanism of lovastatin-induced apoptosis in AML cells. In support of this hypothesis, the geranylgeranyl transferase inhibitor (GGTI-298) mimicked the effect of lovastatin, whereas the farnesyl transferase inhibitor (FTI-277) was much less effective at triggering apoptosis in AML cells. Inhibition of geranylgeranylation was monitored and associated with the apoptotic response induced by lovastatin and GGTI-298 in the AML cells. We conclude that blockage of the mevalonate pathway, particularly inhibition of protein geranylgeranylation holds a critical role in the mechanism of lovastatin-induced apoptosis in AML cells.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Leucemia Mieloide/patologia , Lovastatina/farmacologia , Prenilação de Proteína/efeitos dos fármacos , Doença Aguda , Benzamidas/farmacologia , Nucleotídeos de Desoxiuracil/metabolismo , Fluoresceína-5-Isotiocianato , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Metionina/análogos & derivados , Metionina/farmacologia , Ácido Mevalônico/farmacologia , Fosfatos de Poli-Isoprenil/farmacologia , Sesquiterpenos , Células Tumorais Cultivadas
7.
Exp Neurol ; 170(2): 364-70, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11476602

RESUMO

Although the adult mammalian optic nerve does not regenerate following lesion, in the neonatal rat, retinal ganglion cell (RGC) axons retain the capacity to grow across lesion sites in the brain. Following a brachial lesion at postnatal day 2 (P2), some RGC axons, together with ingrowing cortico-tectal axons, cross the lesion to reinnervate the superior colliculus (SC). Here we use immunohistochemistry to examine expression of the guidance cue ephrin A2 following a brachial lesion. Normal animals show a steady decrease in ephrin A2 immunoreactivity between P5 and P31, with a low rostral to high caudal gradient being evident only at P5. By contrast, after brachial lesion, values are significantly elevated rostrally at P5 and caudally at P12; moreover, a steep rostro-caudal gradient is present at both ages. By P31 values fall to normal levels. Following unilateral enucleation at P2, levels are not significantly different from normal. Our results show that innervation but not denervation triggers increased ephrin A2 expression after a brachial lesion.


Assuntos
Lesões Encefálicas/fisiopatologia , Regeneração Nervosa/fisiologia , Células Ganglionares da Retina/fisiologia , Colículos Superiores/fisiologia , Fatores de Transcrição/análise , Animais , Animais Recém-Nascidos , Axônios/fisiologia , Lesões Encefálicas/patologia , Efrina-A2 , Enucleação Ocular , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Imuno-Histoquímica , Ratos , Ratos Wistar , Células Ganglionares da Retina/citologia , Colículos Superiores/patologia , Colículos Superiores/fisiopatologia , Fatores de Transcrição/genética , Vias Visuais/patologia , Vias Visuais/fisiologia , Vias Visuais/fisiopatologia
8.
Clin Cancer Res ; 7(7): 2067-75, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11448925

RESUMO

The statin family of drugs inhibits 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase, the rate-limiting enzyme of the mevalonate pathway, and is used clinically as a safe and effective approach in the control of hypercholesterolemia. We have shown previously (Dimitroulakos, J., Nohynek, D., Backway, K. L., Hedley, D. W., Yeger, H., Freedman, M. H., Minden, M D., and Penn, L. Z. Increased sensitivity of acute myelogenous leukemias to lovastatin-induced apoptosis: a potential therapeutic approach. Blood, 93: 1308-1318, 1999) that lovastatin, a prototypic member of the statin family, can induce apoptosis of human acute myeloid leukemia (AML) cells in a sensitive and specific manner. In the present study, we evaluated the relative potency and mechanism of action of the newer synthetic statins, fluvastatin, atorvastatin, and cerivastatin, to trigger tumor-specific apoptosis. Cerivastatin is at least 10 times more potent than the other statins at inducing apoptosis in AML cell lines. Cerivastatin-induced apoptosis is reversible with the addition of the immediate product of the HMG-CoA reductase reaction, mevalonate, or with a distal product of the pathway, geranylgeranyl pyrophosphate. This suggests protein geranylgeranylation is an essential downstream component of the mevalonate pathway for cerivastatin similar to lovastatin-induced apoptosis. The enhanced potency of cerivastatin expands the number of AML patient samples as well as the types of malignancies, which respond to statin-induced apoptosis with acute sensitivity. Cells derived from acute lymphocytic leukemia are only weakly sensitive to lovastatin cytotoxicity but show robust response to cerivastatin. Importantly, cerivastatin is not cytotoxic to nontransformed human bone marrow progenitors. These results strongly support the further testing of cerivastatin as a novel anticancer therapeutic alone and in combination with other agents in vivo.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Neoplasias/tratamento farmacológico , Piridinas/farmacologia , Doença Aguda , Atorvastatina , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ácidos Graxos Monoinsaturados/farmacologia , Fluvastatina , Ácidos Heptanoicos/farmacologia , Humanos , Indóis/farmacologia , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/patologia , Neoplasias/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Pirróis/farmacologia , Sensibilidade e Especificidade , Células Tumorais Cultivadas
9.
Diabetologia ; 44(5): 637-45, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11380083

RESUMO

AIMS/HYPOTHESIS: Although the urine flow rate is considerably higher in diabetes mellitus, water reabsorption is greatly increased to concentrate an increased amount of solutes. Our study evaluated the expression of aquaporins and urea transporters, which are essential to the urinary concentration process. METHODS: Northern blot and immunoblot were used to quantify mRNA and proteins for aquaporin-2 (AQP2) as well as urea transporters UT-A1, UT-A2 and UT-B1, in subzones of the renal medulla of rats with streptozotocin-induced diabetes. RESULTS: In these rats, glycaemia, urine flow rate and water reabsorption were respectively fourfold, nine-fold and fourfold those of control rats. The AQP2 protein isoforms were significantly up-regulated in outer and inner medulla. In the base and tip of inner medulla, UT-A1 mRNA was significantly up-regulated (three- and 1.3-fold, respectively) as well as the 117 kD protein (ten- and threefold, respectively) whereas the 97 kD protein was not changed or decreased twofold, respectively. This suggests that, in diabetes, the inner medullary collecting duct is endowed with more UT-A1, especially in its initial part. In the case of mRNA and proteins of UT-A2, located in thin descending limbs in the inner stripe of outer medulla, they were respectively not changed and down-regulated in diabetic rats. CONCLUSION/INTERPRETATION: This study shows that in diabetes, the increased expression of AQP2 and UT-A1 in medullary collecting duct is consistent with an improved concentrating activity. In addition, the underexpression of UT-A2 and the overexpression of UT-A1 in the initial medullary collecting duct are reminiscent of the changes seen after experimental reduction of urine concentration or low protein feeding.


Assuntos
Aquaporinas/genética , Proteínas de Transporte/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatologia , Regulação da Expressão Gênica , Medula Renal/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras , Animais , Aquaporina 2 , Aquaporina 6 , Glicemia/metabolismo , Peso Corporal , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Tipo 1/sangue , Diurese , Glicosúria , Canais Iônicos/genética , Rim/fisiopatologia , Masculino , Tamanho do Órgão , RNA Mensageiro/genética , Ratos , Ratos Wistar , Transcrição Gênica , Ureia/sangue , Ureia/urina , Transportadores de Ureia
10.
Kidney Int ; 58(3): 1202-10, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10972682

RESUMO

BACKGROUND: The facilitated urea transporters (UT), UT-A1, UT-A2, and UT-B1, are involved in intrarenal recycling of urea, an essential feature of the urinary concentrating mechanism, which is impaired in chronic renal failure (CRF). In this study, the expression of these UTs was examined in experimentally induced CRF. METHODS: The abundance of mRNA was measured by Northern analysis and that of corresponding proteins by Western blotting in rats one and five weeks after 5/6 nephrectomy (Nx). RESULTS: At five weeks, urine output was enhanced threefold with a concomitant decrease in urine osmolality. The marked rise in plasma urea concentration and fall in urinary urea concentration resulted in a 30-fold decrease in the urine/plasma (U/P) urea concentration ratio, while the U/P osmoles ratio fell only fourfold. A dramatic decrease in mRNA abundance for the three UTs was observed, bringing their level at five weeks to 1/10th or less of control values. Immunoblotting showed complete disappearance of the 97 and 117 kD bands of UT-A1, and considerable reduction of UT-A2 and UT-B1 in the renal medulla. Similar, but less intense, changes were observed at one-week post-Nx. In addition to the kidney, UT-B1 is also normally expressed in brain and testis. In the brain, its mRNA expression remained normal one-week post-Nx, but decreased to about 30% of normal at five-weeks post-Nx, whereas no change was seen in testis. CONCLUSIONS: (1) The decline in urinary concentrating ability seen in CRF is largely due to a major reduction of UTs involved in the process of urea concentration in the urine, while factors enabling the concentration of other solutes are less intensely affected. (2) The marked reduction of brain UT expression in CRF may be responsible for brain edema of dialysis disequilibrium syndrome observed in some patients after fast dialysis.


Assuntos
Encéfalo/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Medula Renal/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Uremia/metabolismo , Animais , Especificidade de Anticorpos , Northern Blotting , Western Blotting , Proteínas de Transporte/imunologia , Creatinina/sangue , Edema/metabolismo , Expressão Gênica/fisiologia , Capacidade de Concentração Renal/fisiologia , Falência Renal Crônica/metabolismo , Falência Renal Crônica/terapia , Masculino , Glicoproteínas de Membrana/imunologia , Nefrectomia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Diálise Renal , Testículo/metabolismo , Ureia/metabolismo , Uremia/terapia , Transportadores de Ureia
11.
Exp Physiol ; 85 Spec No: 243S-252S, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10795928

RESUMO

Urea is the most abundant urinary solute and is excreted in urine at a much higher concentration than in other body fluids. Urea concentration is achieved in the kidney through complex urea movements between blood vessels and renal tubules, which involve facilitated urea transport. Three major urea transporters expressed in the kidney have been cloned, UT-A1, UT-A2 and UT-B1, the first two derived from the same gene by differential transcription. These membrane proteins enable facilitated diffusion of urea through specific parts of the nephron (UT-A) and through renal vasculature (UT-B) in the medulla. UT-A1 is localised in the terminal part of the inner medullary collecting ducts and accounts for the vasopressin-dependent increase in urea permeability of this segment. UT-A2 is found in the descending thin limbs of Henle's loops. UT-B1 is expressed in the endothelium of the descending vasa recta supplying blood to the renal medulla, and in red cells. All three urea transporters are primarily involved in the process of intrarenal urea recycling, which enables the establishment, and prevents the dissipation, of a high concentration of urea in the inner medulla. This is an essential feature for producing a concentrated urine and thus for water economy in mammals. Vasopressin, upon binding to V2 receptors in the inner medullary collecting ducts, increases urea permeability through activation of UT-A1 molecules, thus enabling urea to diffuse into the inner medullary interstitium. Urea then taken up in ascending vasa recta is returned to the inner medulla via UT-A2 and UT-B1 by countercurrent exchange. These latter two urea transporters are not influenced acutely by vasopressin, but UT-A2 expression is markedly increased in the descending thin limbs of the loops of Henle after sustained exposure to vasopressin or its V2 agonist dDAVP. This effect is indirect because vasopressin receptors are lacking in the descending limbs. The acute direct and delayed indirect actions of vasopressin on renal urea transporters will increase medullary urea accumulation and thus the ability of the kidney to conserve water. Atrial natriuretic peptide inhibits the vasopressin-dependent increase in urea permeability in the inner medullary collecting ducts. The interruption of urea recycling probably contributes to the natriuresis. Impairing in this way the capacity of the kidney to concentrate urea enhances its capacity to concentrate sodium in the urine.


Assuntos
Proteínas de Transporte/metabolismo , Rim/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Ureia/metabolismo , Vasopressinas/metabolismo , Animais , Proteínas de Transporte/química , Rim/química , Glicoproteínas de Membrana/química , Transportadores de Ureia
12.
Biochem Biophys Res Commun ; 271(2): 368-73, 2000 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-10799304

RESUMO

The UT-A2 urea transporter is involved in the recycling of urea through the kidney, a process required to maintain high osmotic gradients. Dehydration increases UT-A2 expression in vivo. The tissue distribution of UT-A2 suggested that hyperosmolarity, and not vasopressin, might mediate this effect. We have analyzed the regulation of UT-A2 expression by ambiant osmolarity both in vitro (mIMCD3 cell line) and in vivo (rat kidney medulla). The UT-A2 mRNA was found to be synergistically up-regulated by a combination of NaCl and urea. Curiously, the UT-A2 protein was undetectable in this hypertonic culture condition, or after transfection of the UT-A2 cDNA, whereas it could be detected in HEK-293 transfected cells. Treating rats with furosemide, a diuretic which decreases the kidney interstitium osmolarity without affecting vasopressin levels, led to decreased levels of the UT-A2 protein. Our results show that the UT-A2 urea transporter is regulated by hyperosmolarity both in vitro and in vivo.


Assuntos
Proteínas de Transporte/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Cloreto de Sódio/farmacologia , Ureia/farmacologia , Animais , Northern Blotting , Western Blotting , Proteínas de Transporte/genética , Linhagem Celular , Diuréticos/farmacologia , Furosemida/farmacologia , Humanos , Medula Renal/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Concentração Osmolar , RNA/metabolismo , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual , Transcrição Gênica , Transfecção , Regulação para Cima , Transportadores de Ureia
13.
J Pept Res ; 55(2): 129-39, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10784029

RESUMO

Salmon calcitonin solutions (50 mg/mL and 100 mg/mL) were placed on stability at 37 degrees C for 1 year in a variety of solvent systems including water, ethanol, glycerol, propylene glycol (PG) and dimethyl sulfoxide (DMSO). Calcitonin degradation was monitored by RP-HPLC and size-exclusion chromatography. DMSO and pH 3.3 solutions provided optimum stability. Conformational stability was also monitored by FTIR over the 1 year time course and compared with chemical and physical stability. After 12 months at 37 degrees C, four major conformations were observed: a beta-sheet conformation (pH 3.3, pH 5.0, 70% DMSO and 70% glycerol), an aggregate conformation (pH 7.0 water), a strong alpha-helical conformation (70% EtOH, 70% PG) and a weak alpha-helical conformation (100% DMSO). No correlation between structure and chemical stability was observed in which both the beta-sheet structure (pH 3.3, water) and a loose alpha-helical structure (100% DMSO) demonstrated good stability. However, some correlation was observed between structure and physical stability, where co-solvents inducing an alpha-helical structure resulted in a decrease in gelation. These two structural states associated with improved stability and minimal gelation, indicated that gelation can be reduced or eliminated by the use of pharmaceutically acceptable co-solvents. Finally, salmon calcitonin (50 mg/mL) was formulated in 100% DMSO and delivered from a DUROS implant over 4 months. Delivery at a target dose of 18 microg/day calcitonin at 37 degrees C was confirmed.


Assuntos
Calcitonina/farmacologia , Implantes de Medicamento/farmacologia , Sequência de Aminoácidos , Animais , Composição de Medicamentos , Estabilidade de Medicamentos , Dados de Sequência Molecular , Osmose , Conformação Proteica , Salmão , Soluções , Espectroscopia de Infravermelho com Transformada de Fourier
14.
Proc Natl Acad Sci U S A ; 96(18): 10397-402, 1999 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-10468619

RESUMO

Diabetic nephropathy represents a major complication of diabetes mellitus (DM), and the origin of this complication is poorly understood. Vasopressin (VP), which is elevated in type I and type II DM, has been shown to increase glomerular filtration rate in normal rats and to contribute to progression of chronic renal failure in 5/6 nephrectomized rats. The present study was thus designed to evaluate whether VP contributes to the renal disorders of DM. Renal function was compared in Brattleboro rats with diabetes insipidus (DI) lacking VP and in normal Long-Evans (LE) rats, with or without streptozotocin-induced DM. Blood and urine were collected after 2 and 4 weeks of DM, and creatinine clearance, urinary glucose and albumin excretion, and kidney weight were measured. Plasma glucose increased 3-fold in DM rats of both strains, but glucose excretion was approximately 40% lower in DI-DM than in LE-DM, suggesting less intense metabolic disorders. Creatinine clearance increased significantly in LE-DM (P < 0.01) but failed to increase in DI-DM. Urinary albumin excretion more than doubled in LE-DM but rose by only 34% in DI-DM rats (P < 0.05). Kidney hypertrophy was also less intense in DI-DM than in LE-DM (P < 0.001). These results suggest that VP plays a critical role in diabetic hyperfiltration and albuminuria induced by DM. This hormone thus seems to be an additional risk factor for diabetic nephropathy and, thus, a potential target for prevention and/or therapeutic intervention.


Assuntos
Albuminúria , Diabetes Insípido/fisiopatologia , Diabetes Mellitus Experimental/fisiopatologia , Nefropatias Diabéticas/fisiopatologia , Taxa de Filtração Glomerular , Falência Renal Crônica/fisiopatologia , Rim/fisiopatologia , Vasopressinas/fisiologia , Animais , Creatinina/metabolismo , Diabetes Insípido/genética , Glicosúria , Hipertrofia , Rim/patologia , Masculino , Nefrectomia , Tamanho do Órgão , Ratos , Ratos Brattleboro , Ratos Long-Evans , Vasopressinas/deficiência , Vasopressinas/genética
15.
J Comp Neurol ; 412(4): 617-32, 1999 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-10464358

RESUMO

This study examined whether prior regenerative growth through peripheral nerve (PN) bridging grafts influenced the specificity with which lesioned adult rat retinal ganglion cell (RGC) axons grew into co-grafts of developing target tissue (fetal superior colliculus). Growth into nontarget (muscle) tissue was also examined. Autologous PN was grafted onto the transected optic nerve. After 14 days, the distal ends of the PNs were placed next to, or inserted into, embryonic tectal tissue or into autologous muscle grafts placed in frontal cortex cavities. Host retinal projections were examined 3-8 months later using anterograde and retrograde tracing techniques. In rats in which there was good apposition between PN and tectal tissue, small numbers of RGC axons were observed growing into the tectal grafts (maximum distance of 180 microm). No evidence of specific innervation of appropriate target regions within tectal grafts was detected, even though such regions (identified by acetylcholinesterase histochemistry) were often located close to the PN grafts. In rats with PN/muscle co-grafts, the extent of retinal axon outgrowth was greater (up to 465 microm from the PN tip) and labelled profiles that resembled motor endplates were seen contacting muscle fibres. Previous studies have shown that spontaneously regenerating RGC axons consistently and selectively innervate appropriate target areas in fetal tectal tissue grafted directly into optic tract lesion cavities. Together, the data suggest that exposure to a PN environment may have reduced the extent of adult retinal axon growth into fetal tectal transplants and affected the way regenerating axons responded to specific developmental cues expressed by target cells in the co-grafted tissue.


Assuntos
Axônios/fisiologia , Músculos/transplante , Regeneração Nervosa , Nervos Periféricos/transplante , Células Ganglionares da Retina/fisiologia , Colículos Superiores/transplante , Animais , Transplante de Tecido Fetal , Imuno-Histoquímica , Ratos , Ratos Wistar , Células Ganglionares da Retina/ultraestrutura , Colículos Superiores/embriologia , Conjugado Aglutinina do Germe de Trigo-Peroxidase do Rábano Silvestre
16.
J Pept Res ; 53(4): 432-41, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10406221

RESUMO

The effect of solvent on the rate of leuprolide degradation and on the structure of the degradation products was explored. Leuprolide solutions (370 mg/mL) were prepared in water and dimethyl sulfoxide (DMSO) for delivery in DUROS osmotic implants. Both solvent systems demonstrated better than 90% stability after 1 year at 37 degrees C, where the DMSO formulation afforded better stability than the aqueous formulation and was used in subsequent clinical trials. The rate of leuprolide degradation in DMSO was also observed to accelerate with increasing moisture content, indicating that the aprotic solvent minimized chemical degradation. Interestingly, leuprolide degradation products varied with formulation vehicle. The proportions of leuprolide degradation products observed to form in water and DMSO at 37 degrees C were hydrolysis > aggregation > isomerization > oxidation and aggregation > oxidation > hydrolysis > isomerization, respectively. Specifically, more N-terminal hydrolysis and acetylation were observed under aqueous conditions, and increased Trp oxidation and Ser beta-elimination were seen under non-aqueous conditions. Furthermore, the major chemical degradation pathway changed with temperature in the DMSO formulation (decreasing oxidation with increasing temperature), but not in the aqueous formulation.


Assuntos
Dimetil Sulfóxido/química , Leuprolida/química , Água/química , Acetilação , Sequência de Aminoácidos , Cromatografia Líquida de Alta Pressão/métodos , Estabilidade de Medicamentos , Hidrólise , Isomerismo , Leuprolida/metabolismo , Espectrometria de Massas , Oxirredução , Estrutura Secundária de Proteína
17.
Exp Nephrol ; 7(1): 44-51, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-9892813

RESUMO

Differences in dietary protein level induce differences in fractional excretion of urea, in arginine vasopressin (AVP) plasma level, and in urine concentrating activity (in which intervene the renal urea transporters (UT)). The abundance of mRNA for UT-A1 (of the inner medullary collecting duct, IMCD) UT-A2 (of the descending thin limb) and UT-B1 (of descending vasa recta) was determined by Northern analysis of total RNA extracted from medullary subregions of Sprague-Dawley rats fed for 1 week, a low, normal, or high protein diet. The implication of AVP was then examined by studying AVP-deprived (Brattleboro) rats. Our results show that none of these transporters is affected by the level of protein intake, except UT-A1 that is reduced in terminal IMCD by low protein diet in the absence of AVP (Brattleboro rats). These data suggest that (1) the previously reported effect of kidney medulla hypertonicity on UT-A2 and UT-B1 mRNA expression is somehow obliterated by protein intake deficiency or excess, and (2) AVP influences the mRNA abundance of the UT-A1 of the terminal IMCD during protein deficiency.


Assuntos
Proteínas de Transporte/genética , Proteínas Alimentares/administração & dosagem , Rim/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ureia/metabolismo , Animais , Arginina Vasopressina/sangue , Arginina Vasopressina/deficiência , Expressão Gênica , Medula Renal/metabolismo , Túbulos Renais Coletores/metabolismo , Masculino , Ratos , Ratos Brattleboro , Ratos Sprague-Dawley , Transportadores de Ureia
18.
Exp Nephrol ; 6(6): 471-9, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9807016

RESUMO

Specific urea transporters are responsible for the rapid urea movements occurring in precise medullary structures of the mammalian kidney. Three of them, ensuring facilitated passive transports, have been cloned yet: UT2-long is responsible for the high vasopressin-dependent urea permeability of the terminal inner medullary collecting ducts; UT2-short is located along a short portion of the thin descending limbs of Henle's loops; UT11 is expressed along the descending vasa recta. These three transporters are involved in the accumulation of urea in the medulla, participating to the corticopapillary osmotic gradient required for urine concentration in the presence of antidiuretic hormone. UT2-long enables diffusion of urea in the inner medulla, and UT2-short and UT11 enable the recycling of this urea by counter-exchange. These transporters could also be involved in nitrogen balance by modulation of their expression according to the need for urea excretion (protein-rich diet), or for nitrogen conservation (protein-poor diet). Several other urea transporters, including active transporters responsible for urea secretion or reabsorption, remain to be cloned and characterized.


Assuntos
Proteínas de Transporte/fisiologia , Rim/metabolismo , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana Transportadoras , Animais , Água Corporal/metabolismo , Nitrogênio/urina , Transportadores de Ureia
19.
Pharm Res ; 15(9): 1442-8, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9755898

RESUMO

PURPOSE: The purpose of this study was to characterize the conformation, aggregation, and stability of leuprolide on gelation. METHODS: Infrared spectra (FTIR) of leuprolide solutions and gels were collected in water, propylene glycol (PG), dimethyl sulfoxide (DMSO), and trifluoroethanol (TFE). Leuprolide solution and gel stability data were obtained by SEC and RP-HPLC. RESULTS: Leuprolide was induced to gel with increasing peptide concentration, introduction of salts, and gentle agitation. Leuprolide dissolved in water (400 mg/ml) demonstrated FTIR spectra consisting of two major bands of equal intensity at 1615 cm(-1) and 1630 cm(-1), similar to inter- and intra-molecular beta-sheet structure in proteins. When samples were gently agitated for 24 hours at 25 degrees C, the formulation was observed to change from a viscous liquid to an opaque gel with a concomitant shift in infrared spectra from the equal intensity bands to mostly 1630 cm(-1), indicating a shift to a preferred beta-sheet structure. Incubation of leuprolide with 20-200 mM salts at 25 degrees C and 37 degrees C also produced gels ranging from clear to cloudy and stringy white precipitates. The gel and precipitate were marked by a shift of the predominant beta-sheet band to 1630 cm(-1) and 1615 cm(-1), respectively. Leuprolide was also observed to gel and/or precipitate in mixtures of water, PG or TFE, but not in DMSO. CONCLUSIONS: Birefringence was noted in many of the firmer gels. Both solutions and gels demonstrated minimal dimer or trimer formation, with no larger order aggregates detected. The chemical stability profile of gelled leuprolide was similar to that of the non-gelled water formulation by RP-HPLC.


Assuntos
Leuprolida/química , Cromatografia Líquida de Alta Pressão , Dimetil Sulfóxido/química , Estabilidade de Medicamentos , Géis , Peptídeos/química , Conformação Proteica , Solventes/química , Espectroscopia de Infravermelho com Transformada de Fourier
20.
J Am Soc Nephrol ; 9(8): 1359-66, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9697657

RESUMO

In the kidney, facilitated urea transport in precise vascular and tubular structures is mainly involved in water conservation. Three urea transporters have been cloned: UT2-long expressed in terminal inner medullary collecting duct (IMCD), UT2-short expressed in thin descending limb, and UT11 in descending vasa recta. The effect of arginine vasopressin (AVP) administration on mRNA expression of these three transporters was examined in Brattleboro rats with diabetes insipidus. V2 effects were discriminated from combined V1 + V2 effects by comparing treatments with 1-deamino-8-D-AVP (dDAVP) (selective V2 agonism) and AVP (V1 and V2 agonism). Acute and chronic treatments were studied. Abundance of specific mRNA was assessed by quantitative Northern blot analysis of RNA extracted from two regions of inner stripe of outer medulla and from two regions of inner medulla (IM). The results show that mRNA of these urea transporters are differently regulated by AVP. (1) Long-term treatment with either AVP or dDAVP does not alter UT2-long mRNA in tip IM (terminal IMCD) except for a transient initial decrease. (2) Unlike AVP, dDAVP induces the appearance of significant expression of UT2-long mRNA in base IM (initial IMCD), indicating a major V2 effect. (3) UT2-short mRNA in deep inner stripe of outer medulla and base IM (thin descending limb of short and long loops, respectively) is progressively upregulated with duration of AVP or dDAVP treatment. (4) The much higher changes in UT2-long and UT2-short induced by dDAVP compared with AVP suggest that they are dependent mainly on V2 agonism, and likely attenuated by V1 agonism. (5) UT11 mRNA expression in tip IM is equally depressed by AVP and dDAVP, indicating that this vascular transporter is also influenced by AVP and/or urine-concentrating activity, via an indirect mechanism that remains to be determined.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Rim/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Ureia/metabolismo , Vasopressinas/farmacologia , Animais , Arginina Vasopressina/farmacologia , Desamino Arginina Vasopressina/farmacologia , Diabetes Insípido/genética , Diabetes Insípido/metabolismo , Expressão Gênica/efeitos dos fármacos , Rim/irrigação sanguínea , Rim/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Brattleboro , Distribuição Tecidual , Transportadores de Ureia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA