Literature
首页医源资料库在线期刊美国生理学杂志2006年第289卷第6期

Regulation of thick ascending limb transport: role of nitric oxide

来源:《美国生理学杂志》
摘要:Inthekidney,NOreducesrenalvascularresistance,increasesglomerularfiltrationrate,altersreninrelease,andinhibitstransportalongthenephron。Thethickascendinglimbisresponsibleforabsorbing20-30%ofthefilteredloadofNaCl,muchofthebicarbonatethatescapestheproximal......

点击显示 收起

【摘要】  Nitric oxide (NO) plays a role in many physiological and pathophysiological processes. In the kidney, NO reduces renal vascular resistance, increases glomerular filtration rate, alters renin release, and inhibits transport along the nephron. The thick ascending limb is responsible for absorbing 20-30% of the filtered load of NaCl, much of the bicarbonate that escapes the proximal nephron, and a significant fraction of the divalent cations reclaimed from the forming urine. Additionally, this nephron segment plays a role in K + homeostasis. This article will review recent advances in our understanding of the role NO plays in regulating the transport processes of the thick ascending limb. NO has been shown to inhibit NaCl absorption primarily by reducing Na + -K + -2Cl - cotransport activity. NO also inhibits bicarbonate absorption by reducing Na + /H + exchange activity. It has also been reported to enhance luminal K + channel activity and thus is likely to alter K + secretion. The source of NO may be vascular structures such as the afferent arteriole or vasa recta, or the thick ascending limb itself. NO is produced by NO synthase 3 in this segment, and several factors that regulate its activity both acutely and chronically have recently been identified. Although the effects of NO on thick ascending limb transport have received a great deal of attention recently, its effects on divalent ion absorption and many other issues remain unexplored.

【关键词】  sodiumpotassium chloride cotransport sodium/hydrogen exchanger cGMP bicarbonate calcium magnesium Na + K + ATPase


NITRIC OXIDE ( NO ) IS A KEY regulator of physiological processes, ranging from long-term potentiation of synaptic transmission in the brain ( 7, 9, 86 ) to dilatation of resistance vessels in the circulatory system ( 14, 43, 44 ). In the kidney, NO reduces renal vascular resistance ( 64, 87 ), increases glomerular filtration rate ( 5, 45, 46, 60, 80 ), and appears to have a biphasic effect on renin release ( 48, 82, 91 ). NO also regulates solute and solvent transport along the nephron ( 71 ). The importance of NO as a regulator of ion and water transport was first demonstrated by in vivo experiments showing that stimulating NO production increased urinary Na + and water excretion ( 6, 50, 62, 96 ), whereas inhibiting NO generation decreased excretion ( 49 - 51 ). In many situations, the natriuretic and diuretic effects of NO were not accompanied by changes in glomerular filtration rate or renal blood flow ( 58, 59 ), suggesting that NO directly inhibits Na + and water transport along the nephron. The source of NO in these early studies was not investigated and could have involved one or more of the three NO synthase (NOS) isoforms expressed in the kidney: NOS1, NOS2, and NOS3. Originally, these enzymes were referred to as inducible (iNOS), neuronal (nNOS), and endothelial (eNOS) after the tissues where they were identified; but this nomenclature now creates confusion, as each NOS isoform is expressed in many cell types ( 37 ). The thick ascending limb of the loop of Henle absorbs 20-30% of the filtered load of NaCl ( 27 ), and because it is water impermeable it generates most of the osmotic gradient that drives fluid absorption in the collecting duct system. Thus it is logical to assume that some of the effects of NO on urinary Na + excretion and urinary volume are due to its actions in the thick ascending limb.


TRANSPORTERS INVOLVED IN NaCl ABSORPTION


NaCl absorption in the thick ascending limb occurs via both transcellular and paracellular pathways. Transcellular absorption is an active process in which Na + and Cl - enter thick ascending limb cells via luminal Na + -K + -2Cl - cotransporters. Cl - ions exit across the basolateral plasma membrane via Cl - channels and/or K + -Cl - cotransport ( 26 ). The basolateral Na + -K + -ATPase generates the driving force for NaCl entry by extruding Na + across the basolateral membrane ( 63 ). Na + is also absorbed via the paracellular pathway due to the lumen-positive voltage caused by recycling of K + across the luminal membrane and back into the forming urine ( 29 - 34 ).


The effects of NO on NaCl absorption were first demonstrated in isolated, perfused tubules. NO from a donor was found to inhibit Cl - flux ( J Cl ) ( 78 ). These data indicated that NO from other sources, such as the afferent arterioles, descending vasa recta, or other nephron segments, may regulate NaCl transport in the thick ascending limb via generation and diffusion of NO. Addition of L -arginine, the substrate of NOS, also decreased J Cl, and this effect was blocked by the NOS inhibitor N G -nitro- L -arginine methyl ester ( L -NAME). These data suggest that NO is produced by thick ascending limbs and acts as an autacoid to inhibit NaCl absorption ( 78 ). The conclusion that NO is produced by the thick ascending limb was confirmed by direct measurements of NO in isolated, perfused thick ascending limbs as well as in thick ascending limb suspensions using two different techniques: a NO-specific fluorescent dye ( 67, 85 ) and a NO-selective electrode ( 38, 73 ).


The inhibitory effects of NO on thick ascending limb J Cl could be due to inhibition of one of the luminal membrane transporters (Na + -K + -2Cl - cotransporter or K + channels) or one of the basolateral transporters (Na + -K + -ATPase; Cl - channels or K + -Cl - cotransporter) ( Fig. 1 ). Data showing that NO decreased both intracellular Na + and Cl - indicate that NO decreases NaCl entry rather than exit. Furthermore, because the Na + -K + -2Cl - cotransporter links Na + and Cl - entry, the effect of NO on J Cl was most likely due to inhibition of the luminal Na + -K + -2Cl - cotransporter. Studies directly investigating Na + -K + -2Cl - cotransport activity confirmed this. Measurements of initial Na + influx when luminal NaCl was increased from 0 to 130 mM in isolated, perfused thick ascending limbs revealed that the rate was lower after NO treatment ( 72 ).


Fig. 1. Direct effects of nitric oxide (NO) on individual transporters of the thick ascending limb (THAL) cell. NO directly inhibits Na + -K + -2Cl - cotransport and Na/H exchangers (NHE1/3). NO inhibits the Na + -K + -ATPase pump by reacting with O 2 - and forming OONO -. The effect of NO on inwardly rectified (ROMK) channels remains controversial. (-), Inhibition; (?), controversial; NKCC2, Na + -K + -2Cl - cotransporter; NHE3, apical Na + /H + exchanger; NHE1, basolateral Na + /H + exchanger.


The direct measurements of inhibition of Na + -K + -2Cl - cotransport by NO conflict with the predicted effect based on patch-clamp studies of luminal K + channels. In patch-clamp studies, NO donors have been reported to stimulate the inwardly rectified K + channel (ROMK) ( 57 ). This led to the prediction that NO would stimulate Na + -K + -2Cl - cotransport in the thick ascending limb and thus NaCl absorption, because inhibiting ROMK reduces Na + -K + -2Cl - cotransport ( 40 ). However, this reasoning was proven incorrect, because increasing luminal K + depolarized the luminal membrane potential to the same extent before and after treatment of isolated, perfused tubules with NO, suggesting that NO did not have a significant effect on luminal K + permeability. Additionally, increasing luminal membrane K + permeability with valinomycin did not block the inhibitory action of NO ( 72 ). Why patch-clamp studies show that NO stimulates ROMK, whereas other studies do not remains an unanswered question.


Although NO reduces intracellular Na + and Cl - and inhibits Na + -K + -2Cl - cotransport, this does not necessarily indicate that NO has no effect on Na + -K + -ATPase, only that the magnitude of the effect is relatively less than its effect on the cotransporter. However, at least acutely, NO does not appear to have an effect on Na + -K + -ATPase activity. In suspensions of thick ascending limbs, in the presence of 10.5 mM extracellular Na +, NO inhibited oxygen consumption to the same extent as furosemide. Addition of nystatin to equilibrate extra- and intracellular Na + returned oxygen consumption to control levels in both NO- and furosemide-treated tubules. These data indicate that NO does not affect the affinity constant of Na + -K + -ATPase for intracellular Na +. Additionally, nystatin stimulated oxygen consumption in NO- and furosemide-treated suspensions to the same extent in the presence of 145.5 mM Na +. These data indicate that NO does not affect the maximum rate of Na + -K + -ATPase ( 72 ).


Although NO does not appear to inhibit Na + -K + -ATPase acutely, this may not be the case chronically. After exposure of thick ascending limbs to a NO donor for 2 h, pump activity was reduced by 32% ( 90 ), similar to cultured mouse proximal tubule cells ( 28 ). Inhibition in both studies depended on superoxide, because superoxide scavengers abolished the inhibitory effects of NO on Na + -K + -ATPase activity. These results indicate that OONO - rather than NO per se was responsible. This raises the possibility that inhibition of Na + -K + -ATPase by OONO - is an artifact of the experimental design due to nonphysiological P O 2 rather than a physiologically significant regulatory process. P O 2 values of the outer medulla are 10-20 mmHg ( 94 ) and even those of the cortex are only 40 mmHg ( 93 ), whereas P O 2 in both studies was 160 mmHg, the same as room air.


Besides the potential chronic effects of NO on Na + -K + -ATPase, Turban et al. ( 88 ) reported that systemic blockade of NOS by infusion of L -NAME decreased the renal abundance of Na + -K + -2Cl - cotransporters during aldosterone escape, suggesting that NO stimulates thick ascending limb cotransporter expression. However, this observation could have been the result of the increased blood pressure rather than a direct effect of NO on cotransporter expression.


The signaling cascade whereby NO inhibits the Na + -K + -2Cl - cotransporter involves a decrease in cAMP ( 70 ), as does inhibition of vasopressin-stimulated osmotic water permeability in the collecting duct ( 18 ). NO increases cGMP by stimulating soluble guanylate cyclase. In turn, cGMP activates cGMP-stimulated phosphodiesterase (phosphodiesterase II). This enhances the degradation of cAMP and thus reduces Na + -K + -2Cl - activity ( 70 ). Because cAMP stimulates the Na + -K + -2Cl - cotransporter and has recently been shown to increase membrane expression of Na + -K + -2Cl - cotransporters ( 68 ), the mechanism whereby NO decreases Na + -K + -2Cl - cotransport activity is likely to be mediated by reducing cotransporter levels at the apical membrane. However, such a mechanism has not yet been elucidated to our knowledge.


The effects of NO on the other basolateral transporters involved directly or indirectly with NaCl absorption have not been studied. These include basolateral K + channels, Cl - channels, and K + -Cl - cotransporters.


TRANSPORTERS INVOLVED IN ACID-BASE BALANCE


The thick ascending limb absorbs most of the HCO 3 - that escapes the proximal tubule ( 21 ). HCO 3 - absorption is due to secretion of H + via luminal Na + /H + exchange. Consequently, HCO 3 - reabsorption is regulated primarily through this apical exchanger ( 21, 22, 24 ). HCO 3 - transport across the basolateral membrane occurs via basolateral Na + /HCO 3 - (NBCn1) ( 65 ), Cl - /HCO 3 - (AE 2 ) ( 79 ), and K + /HCO 3 - exchangers ( 8 ).


Direct measurements of HCO 3 - flux in the isolated, perfused tubule demonstrated that stimulation of endogenous NO production by addition of the NOS substrate L -arginine inhibited thick ascending limb HCO 3 - transport ( 69 ). This effect could be explained by direct inhibition of apical Na + /H + exchanger activity, blockade of the basolateral HCO 3 - cotransporter or a decrease in basolateral Na + -K + -ATPase activity, leading to a diminished electrochemical gradient across the membrane. Measurements of the effects of NO on steady-state and intracellular pH (pH i ) recovery after an acid load demonstrated that the NO donors spermine-NONOate and nitroglycerin inhibited both steady-state pH i and proton efflux as calculated from pH i recovery experiments in isolated, perfused thick ascending limbs ( 20 ). Because NO has no direct acute effect on Na + -K + -ATPase ( 72 ), a decrease in Na + /H + exchange can only be explained by a direct effect of NO on the exchanger. In addition, by selectively inhibiting the basolateral Na + /H + exchanger with dimethyl amiloride, it was found that NO directly inhibited luminal Na + /H + exchange. Although the inhibitory effect of NO on thick ascending limb HCO 3 - could also be explained by inhibition of basolateral transporters, the direct effects of NO on basolateral HCO 3 - transporters have not been investigated to our knowledge.


The signaling cascade whereby NO inhibits thick ascending limb HCO 3 - absorption is different from that responsible for inhibition of NaCl absorption. The inhibitory effect of endogenous NO can be mimicked by the cell-permeant cGMP analogs, dibutyryl cGMP and 8-bromoadenosine-cGMP. Addition of L -arginine did not change the rate of HCO 3 - absorption after pretreatment with dibutyryl cGMP. These data indicate that cGMP mediates all of the effects of NO on HCO 3 - reabsorption by this segment. In addition, the inhibitory effect of L -arginine on HCO 3 - transport was completely blocked by a cGMP-dependent protein kinase inhibitor ( 69 ).


In contrast to these data, Good et al. ( 23 ) described a stimulatory effect of L -arginine and NO donors on thick ascending limb HCO 3 - transport. Although the explanation for such disparate results is unclear, it may be due to the age of the animals or different diets. The rats Good et al. used were 2-3 wk younger than those in the study showing NO-induced inhibition ( 18 ) and were fed a different diet. Both age and diet could alter endogenous cyclic nucleotide levels and thus the effect of NO ( 19, 56 ).


In addition to absorbing HCO 3 -, the thick ascending limb has been reported to secrete this ion via a recently identified K + -dependent cotransporter at the apical membrane ( 92 ). Although we could find no reports of the effects of NO on this transporter, its stimulation would appear as inhibition of net absorption, and differential expression of the K + /HCO 3 - cotransporter could explain the controversy over whether NO stimulates or inhibits HCO 3 - absorption.


The thick ascending limb expresses at least two other transporters involved in acid-base balance: the basolateral Na + /H + exchanger ( 11, 66 ) and a putative K + /NH 4 + exchanger ( 3 ). NO has been shown to inhibit basolateral Na + /H + exchange ( 20 ), but whether NO inhibits or stimulates K + /NH 4 + exchange is unknown.


Besides the acute effects of NO on Na + /H + exchange, very little is known about its chronic effects. Turban et al. ( 88 ) reported that systemic blockade of NOS by infusion of L -NAME decreased renal abundance of Na + /H + exchanger NHE3 during aldosterone escape. These data suggest that NO stimulates thick ascending limb exchanger expression. However, this observation could have been the result of increased blood pressure rather than a direct effect of NO on cotransporter expression.


OTHER IONS


The direct effects of NO on thick ascending limb Mg 2+, Ca 2+, and NH 4 + transport have not been investigated to our knowledge. In this segment, Mg 2+ and Ca 2+ are reabsorbed via the paracellular pathway due to the lumen-positive potential ( 25 ). Thus one would predict that NO would inhibit divalent cation absorption due to its inhibitory effect on Na + -K + -2Cl - cotransport. Similarly, one would expect that NO would diminish NH 4 + absorption, because NH 4 + is absorbed primarily via Na + -K + -2Cl - cotransport. However, there is no direct evidence in the literature addressing either issue.


ACUTE AND CHRONIC REGULATION OF NO PRODUCTION


As discussed above, NO produced by the thick ascending limb acts as an autacoid to inhibit NaCl absorption. Thus a discussion of the factors that regulate NO production is relevant when one is discussing how NO regulates thick ascending limb transport. The thick ascending limb expresses all three NOS isoforms: NOS1, NOS2, and NOS3 ( 4, 36, 52, 61, 67, 84, 89 ). However, studies using knockout mice demonstrated that L -arginine inhibited NaCl reabsorption in thick ascending limbs isolated from NOS1 and NOS2 but not NOS3 knockouts. Furthermore, in vivo gene transfer of NOS3 in the thick ascending limbs of NOS3 knockout mice restored the inhibitory effect of L -arginine on NaCl reabsorption. These data indicate that NOS3-derived NO is responsible for inhibition of NaCl absorption in the thick ascending limb ( 74, 77 ). However, they do not rule out the possibility that other NOS isoforms regulate other transport processes in the thick ascending limb, or that they play a role in NaCl absorption under different experimental conditions (i.e., placing animals on a high-salt diet or inducing hypertension or diabetes).


The fact that NOS3 is responsible for the inhibition of thick ascending limb NaCl absorption begs the question of whether NOS3 in the thick ascending limb is regulated by the same factors that stimulate its activity in endothelial cells. In endothelial cells, endothelin-1 ( 12, 39 ), 2 -adrenergic activation ( 1 ), and ATP ( 47 ) all stimulate NOS3 to produce NO. In the isolated, perfused thick ascending limb, we ( 76 ) and others ( 42 ) found that endothelin-1 inhibited NaCl reabsorption. This inhibitory effect was blunted by the NOS inhibitor L -NAME and depended on L -arginine. Endothelin-1-dependent inhibition of NaCl absorption was abolished in the presence of an ET B -receptor antagonist and mimicked by an ET B -receptor agonist. In contrast, an ET A antagonist had no effect. Thus endothelin-1 regulates NO production in the thick ascending limb ( 76 ). The 2 -adrenergic agonist clonidine also reduced NaCl reabsorption in the thick ascending limb. Similar to endothelin-1, this inhibition was abolished by the NOS inhibitor L -NAME or removal of L -arginine. These data suggest that 2 -adrenergic receptor activation also regulates NOS in the thick ascending limb ( 75 ). Recent data show that ATP can acutely stimulate NO production in isolated thick ascending limbs by activating P2X receptors ( 85 ). These data suggest that extracellular ATP may regulate NOS3 activity in the thick ascending limb; however, the effects of ATP on NaCl transport have not been reported to our knowledge. In addition to the physiological regulators described above, the antimalarial drug sodium artesunate inhibits NaCl reabsorption in isolated perfused thick ascending limbs ( 10 ). The inhibitory effect on transport was blocked by L -NAME, suggesting that the effect of this drug on renal function may be due to enhanced NO levels in the thick ascending limb.


In addition to humoral factors, mechanical stimuli can also activate NOS3. In endothelial cells, flow activates NOS3 ( 16, 17, 81 ). In isolated, perfused thick ascending limbs, increasing luminal flow from 0 to 20 nl/min stimulated NOS3 activity and NO production ( 73 ). Although blockade of phosphatidylinositol 3-kinase (PI3-kinase) prevented flow-induced NOS3 activation in thick ascending limbs, similar to endothelial cells, activation of NOS3 in the thick ascending limb causes its translocation from intracellular compartments to the luminal membrane. This does not occur in endothelial cells. Given that flow through the thick ascending limb changes both acutely ( 13, 41, 53 - 55, 83 ) and chronically ( 2, 15, 54 ), flow-induced NOS3 activation is likely to play a role in the regulation of NO levels and thick ascending limb transport. However, how NO regulates thick ascending limb NaCl transport under different flow conditions remains to be explored, and our laboratory is currently investigating this.


Although acute regulation of NOS3 activity in the thick ascending limb is similar to endothelial cells in many respects, chronic NOS3 regulation is not. A high-salt diet was shown to augment NOS3 expression in the thick ascending limb. However, this increase in expression was transient, reaching a maximum at 3 days and returning to control levels by 28 days ( 38 ). The effects of high salt on NOS3 expression are mediated by activation of ET B receptors and PI3-kinase ( 35 ) ( Fig. 2 ). A dual ET A/B receptor antagonist blocked the effect of high salt in vivo, and the ability of endothelin to enhance expression was blocked by an ET B -receptor antagonist ( 36 ). Additionally, an inhibitor of PI3-kinase blocked the effects of endothelin on NOS3 expression in culture ( 35 ). An increase in medullary osmolality is also involved. High salt increased outer medullary osmolality, which, in turn, enhanced endothelin release from thick ascending limb cells and stimulated NOS3 expression in primary cultures ( 36 ). The signaling cascade responsible for the decline in NOS 3 expression after longer exposure to a high-salt diet has not been investigated to our knowledge.


Fig. 2. Effects of a high-salt diet on THAL NO synthase 3 (NOS3). A high-salt diet stimulates THAL NOS3 by increasing medullary osmolality and release of endothelin-1 (ET-1), which, in turn, activates ET B receptors and stimulates NOS3. NOS3-derived NO inhibits Na + -K + -2Cl - cotransport, blunting NaCl reabsorption. PI3-K, phosphatidylinositol 3-kinase.


Although the effects of high salt on both NOS3 expression and activity are biphasic, they are not temporally correlated. Expression peaks at 3 days, whereas NO production peaks at 1 day and returns to control levels by 3 days. This dissociation is a result of differential NOS3 phosphorylation ( 38 ). Interestingly, despite unchanged NO levels, inhibition of thick ascending limb NaCl absorption by a given amount of NO was greater in rats fed high salt for 7 days ( 67 ). Thus it appears that the signaling cascade beyond NO is sensitized by high salt, so that equal amounts of NO will have a greater effect. From these data, one can speculate that during the early phase of a high-salt diet ( day 1 ), NOS3 expression and NO production will be elevated and thus NaCl reabsorption will be inhibited. By 7 days, the NO signaling cascade becomes sensitized, so that enhanced NO levels are no longer needed. This could be a protective mechanism against the detrimental effects caused by high levels of NO. Our laboratory is currently investigating the mechanism involved.


NOS3 activity in the thick ascending limb also appears to change as a result of diabetes ( 52 ). Renal medullary NOS3 activity is increased in diabetic rats. Changes in activity were found to correlate with changes in NOS3 phosphorylation ( 95 ).


In summary, endogenous and exogenous NO inhibit thick ascending limb transport by directly acting on individual transporters, and thus NO plays a role in the regulation of Na +, water, and acid-base homeostasis. NO production by the thick ascending limb can be acutely regulated by humoral and mechanical stimuli. Changes in the effects and production of NO occur during physiological stress and pathophysiological conditions. Our understanding of how NO production is regulated in the thick ascending limb, as well as how that process affects transport, is still at a rudimentary level.


ACKNOWLEDGMENTS


This work was supported in part by National Heart, Lung, and Blood Institute Grants HL-28982 and HL-70985 (to J. L. Garvin), SDG Grant 0430031N to P. Ortiz from the American Heart Association, and a predoctoral fellowship (0415404Z) to M. Herrera from the American Heart Association.

【参考文献】
  Angus JA, Cocks TM, and Satoh K. The adrenoceptors on endothelial cells. Fed Proc 45: 2355-2359, 1986.

Arendshorst WJ and Beierwaltes WH. Renal tubular reabsorption in spontaneously hypertensive rats. Am J Physiol Renal Fluid Electrolyte Physiol 237: F38-F47, 1979.

Attmane-Elakeb A, Amlal H, and Bichara M. Ammonium carriers in medullary thick ascending limb. Am J Physiol Renal Physiol 280: F1-F9, 2001.

Bachmann S, Bosse HM, and Mundel P. Topography of nitric oxide synthesis by localizing constitutive NO synthases in mammalian kidney. Am J Physiol Renal Fluid Electrolyte Physiol 268: F885-F898, 1995.

Bachmann S and Mundel P. Nitric oxide in the kidney: synthesis, localization, and function. Am J Kidney Dis 24: 112-129, 1994.

Baer PG, Navar LG, and Guyton AC. Renal autoregulation, filtration rate, and electrolyte excretion during vasodilatation. Am J Physiol 219: 619-625, 1970.

Bon CL and Garthwaite J. On the role of nitric oxide in hippocampal long-term potentiation. J Neurosci 23: 1941-1948, 2003.

Bourgeois S, Masse S, Paillard M, and Houillier P. Basolateral membrane Cl - -, Na + -, and K + -coupled base transport mechanisms in rat MTALH. Am J Physiol Renal Physiol 282: F655-F668, 2002.

Bredt DS. Endogenous nitric oxide synthesis: biological functions and pathophysiology. Free Radic Res 31: 577-596, 1999.

Campos SB, Rouch LH, and Seguro AC. Effects of sodium artesunate, a new antimalarial drug, on renal function. Kidney Int 59: 1044-1051, 2001.

Demaurex N and Grinstein S. Na + /H + antiport: modulation by ATP and role in cell volume regulation. J Exp Biol 196: 389-404, 1994.

De Nucci G, Thomas R, D'Orleans-Juste P, Antunes E, Walder C, Warner TD, and Vane JR. Pressor effects of circulating endothelin are limited by its removal in the pulmonary circulation and by the release of prostacyclin and endothelium-derived relaxing factor. Proc Natl Acad Sci USA 85: 9797-9800, 1988.

Dwyer TM and Schmidt-Nielsen B. The renal pelvis: machinery that concentrates urine in the papilla. News Physiol Sci 18: 1-6, 2003.

Edwards RM and Trizna W. Modulation of glomerular arteriolar tone by nitric oxide synthase inhibitors. J Am Soc Nephrol 4: 1127-1132, 1993.

Ellison DH, Velazquez H, and Wright FS. Adaptation of the distal convoluted tubule of the rat. Structural and functional effects of dietary salt intake and chronic diuretic infusion. J Clin Invest 83: 113-126, 1989.

Fisslthaler B, Dimmeler S, Hermann C, Busse R, and Fleming I. Phosphorylation and activation of the endothelial nitric oxide synthase by fluid shear stress. Acta Physiol Scand 168: 81-88, 2000.

Gallis B, Corthals GL, Goodlett DR, Ueba H, Kim F, Presnell SR, Figeys D, Harrison DG, Berk BC, Aebersold R, and Corson MA. Identification of flow-dependent endothelial nitric-oxide synthase phosphorylation sites by mass spectrometry and regulation of phosphorylation and nitric oxide production by the phosphatidylinositol 3-kinase inhibitor LY294002. J Biol Chem 274: 30101-30108, 1999.

Garcia NH, Stoos BA, Carretero OA, and Garvin JL. Mechanism of the nitric oxide-induced blockade of collecting duct water permeability. Hypertension 27: 679-683, 1996.

Garvin JL and Beierwaltes WH. Response of proximal tubules to angiotensin II changes during maturation. Hypertension 31: 415-420, 1998.

Garvin JL and Hong NJ. Nitric oxide inhibits sodium/hydrogen exchange activity in the thick ascending limb. Am J Physiol Renal Physiol 277: F377-F382, 1999.

Good DW. The thick ascending limb as a site of renal bicarbonate reabsorption. Semin Nephrol 13: 225-235, 1993.

Good DW. Hyperosmolality inhibits bicarbonate absorption in rat medullary thick ascending limb via a protein-tyrosine kinase-dependent pathway. J Biol Chem 270: 9883-9889, 1995.

Good DW, George T, and Wang DH. Angiotensin II inhibits HCO 3 - absorption via a cytochrome P -450-dependent pathway in MTAL. Am J Physiol Renal Physiol 276: F726-F736, 1999.

Good DW and Watts BA III. Functional roles of apical membrane Na + /H + exchange in rat medullary thick ascending limb. Am J Physiol Renal Fluid Electrolyte Physiol 270: F691-F699, 1996.

Greger R. Ion transport mechanisms in thick ascending limb of Henle's loop of mammalian nephron. Physiol Rev 65: 760-797, 1985.

Greger R and Schlatter E. Properties of the basolateral membrane of the cortical thick ascending limb of Henle's loop of rabbit kidney. A model for secondary active chloride transport. Pflügers Arch 396: 325-334, 1983.

Greger R and Velazquez H. The cortical thick ascending limb and early distal convoluted tubule in the urinary concentrating mechanism. Kidney Int 31: 590-596, 1987.

Guzman NJ, Fang MZ, Tang SS, Ingelfinger JR, and Garg LC. Autocrine inhibition of Na + /K + -ATPase by nitric oxide in mouse proximal tubule epithelial cells. J Clin Invest 95: 2083-2088, 1995.

Hebert SC and Andreoli TE. Control of NaCl transport in the thick ascending limb. Am J Physiol Renal Fluid Electrolyte Physiol 246: F745-F756, 1984.

Hebert SC and Andreoli TE. Effects of antidiuretic hormone on cellular conductive pathways in mouse medullary thick ascending limbs of Henle. II. Determinants of the ADH-mediated increases in transepithelial voltage and in net Cl - absorption. J Membr Biol 80: 221-233, 1984.

Hebert SC and Andreoli TE. Ionic conductance pathways in the mouse medullary thick ascending limb of Henle. The paracellular pathway and electrogenic Cl - absorption. J Gen Physiol 87: 567-590, 1986.

Hebert SC, Culpepper RM, and Andreoli TE. NaCl transport in mouse medullary thick ascending limbs. I. Functional nephron heterogeneity and ADH-stimulated NaCl cotransport. Am J Physiol Renal Fluid Electrolyte Physiol 241: F412-F431, 1981.

Hebert SC, Friedman PA, and Andreoli TE. Effects of antidiuretic hormone on cellular conductive pathways in mouse medullary thick ascending limbs of Henle. I. ADH increases transcellular conductance pathways. J Membr Biol 80: 201-219, 1984.

Hebert SC, Reeves WB, Molony DA, and Andreoli TE. The medullary thick limb: function and modulation of the single-effect multiplier. Kidney Int 31: 580-589, 1987.

Herrera M and Garvin JL. Endothelin stimulates endothelial nitric oxide synthase expression in the thick ascending limb. Am J Physiol Renal Physiol 287: F231-F235, 2004.

Herrera M and Garvin JL. A high-salt diet stimulates thick ascending limb eNOS expression by raising medullary osmolality and increasing release of endothelin-1. Am J Physiol Renal Physiol 288: F58-F64, 2005.

Herrera M and Garvin JL. Recent advances in the regulation of nitric oxide in the kidney. Hypertension 45: 1062-1067, 2005.

Herrera M, Silva G, and Garvin JL. A high-salt diet dissociates NO synthase-3 expression and NO production by the THAL. Hypertension 47: 95-101, 2006.

Hirata Y, Hayakawa H, Suzuki E, Kimura K, Kikuchi K, Nagano T, Hirobe M, and Omata M. Direct measurements of endothelium-derived nitric oxide release by stimulation of endothelin receptors in rat kidney and its alteration in salt-induced hypertension. Circulation 91: 1229-1235, 1995.

Huang DY, Osswald H, and Vallon V. Sodium reabsorption in thick ascending limb of Henle's loop: effect of potassium channel blockade in vivo. Br J Pharmacol 130: 1255-1262, 2000.

Ichihara A and Navar LG. Neuronal NOS contributes to biphasic autoregulatory response during enhanced TGF activity. Am J Physiol Renal Physiol 277: F113-F120, 1999.

Jesus Ferreira MC and Bailly C. Luminal and basolateral endothelin inhibit chloride reabsorption in the mouse thick ascending limb via a Ca 2+ -independent pathway. J Physiol 505: 749-758, 1997.

Juncos LA, Garvin J, Carretero OA, and Ito S. Flow modulates myogenic responses in isolated microperfused rabbit afferent arterioles via endothelium-derived nitric oxide. J Clin Invest 95: 2741-2748, 1995.

Juncos LA, Ito S, Carretero OA, and Garvin JL. Removal of endothelium-dependent relaxation by antibody and complement in afferent arterioles. Hypertension 23: I54-I59, 1994.

Kakoki M, Zou AP, and Mattson DL. The influence of nitric oxide synthase 1 on blood flow and interstitial nitric oxide in the kidney. Am J Physiol Regul Integr Comp Physiol 281: R91-R97, 2001.

Kone BC and Baylis C. Biosynthesis and homeostatic roles of nitric oxide in the normal kidney. Am J Physiol Renal Physiol 272: F561-F578, 1997.

Korenaga R, Ando J, Tsuboi H, Yang W, Sakuma I, Toyo-oka T, and Kamiya A. Laminar flow stimulates ATP- and shear stress-dependent nitric oxide production in cultured bovine endothelial cells. Biochem Biophys Res Commun 198: 213-219, 1994.

Kurtz A and Wagner C. Role of nitric oxide in the control of renin secretion. Am J Physiol Renal Physiol 275: F849-F862, 1998.

Lahera V, Navarro J, Biondi ML, Ruilope LM, and Romero JC. Exogenous cGMP prevents decrease in diuresis and natriuresis induced by inhibition of NO synthesis. Am J Physiol Renal Fluid Electrolyte Physiol 264: F344-F347, 1993.

Lahera V, Salom MG, Fiksen-Olsen MJ, Raij L, and Romero JC. Effects of N G -monomethyl- L -arginine and L -arginine on acetylcholine renal response. Hypertension 15: 659-663, 1990.

Lahera V, Salom MG, Miranda-Guardiola F, Moncada S, and Romero JC. Effects of N G -nitro- L -arginine methyl ester on renal function and blood pressure. Am J Physiol Renal Fluid Electrolyte Physiol 261: F1033-F1037, 1991.

Lee DL, Sasser JM, Hobbs JL, Boriskie A, Pollock DM, Carmines PK, and Pollock JS. Posttranslational regulation of NO synthase activity in the renal medulla of diabetic rats. Am J Physiol Renal Physiol 288: F82-F90, 2005.

Letsou GV, Rosales O, Maitz S, Vogt A, and Sumpio BE. Stimulation of adenylate cyclase activity in cultured endothelial cells subjected to cyclic stretch. J Cardiovasc Surg (Torino) 31: 634-639, 1990.

Leyssac PP and Holstein-Rathlou NH. Effects of various transport inhibitors on oscillating TGF pressure responses in the rat. Pflügers Arch 407: 285-291, 1986.

Leyssac PP, Karlsen FM, Holstein-Rathlou NH, and Skøtt O. On determinants of glomerular filtration rate after inhibition of proximal tubular reabsorption. Am J Physiol Regul Integr Comp Physiol 266: R1544-R1550, 1994.

Li XX, Albrecht FE, Robillard JE, Eisner GM, and Jose PA. G regulation of Na/H exchanger-3 activity in rat renal proximal tubules during development. Am J Physiol Regul Integr Comp Physiol 278: R931-R936, 2000.

Lu M, Wang X, and Wang W. Nitric oxide increases the activity of the apical 70-pS K + channel in TAL of rat kidney. Am J Physiol Renal Physiol 274: F946-F950, 1998.

Majid DS, Omoro SA, Chin SY, and Navar LG. Intrarenal nitric oxide activity and pressure natriuresis in anesthetized dogs. Hypertension 32: 266-272, 1998.

Majid DS, Williams A, Kadowitz PJ, and Navar LG. Renal responses to intra-arterial administration of nitric oxide donor in dogs. Hypertension 22: 535-541, 1993.

Manning RD Jr, Hu L, Tan DY, and Meng S. Role of abnormal nitric oxide systems in salt-sensitive hypertension. Am J Hypertens 14: 68S-73S, 2001.

Mattson DL and Higgins DJ. Influence of dietary sodium intake on renal medullary nitric oxide synthase. Hypertension 27: 688-692, 1996.

Mattson DL and Wu F. Control of arterial blood pressure and renal sodium excretion by nitric oxide synthase in the renal medulla. Acta Physiol Scand 168: 149-154, 2000.

Molony DA, Reeves WB, and Andreoli TE. Na + :K + :2Cl - cotransport and the thick ascending limb. Kidney Int 36: 418-426, 1989.

Navar LG, Inscho EW, Majid SA, Imig JD, Harrison-Bernard LM, and Mitchell KD. Paracrine regulation of the renal microcirculation. Physiol Rev 76: 425-536, 1996.

Odgaard E, Jakobsen JK, Frische S, Praetorius J, Nielsen S, Aalkjaer C, and Leipziger J. Basolateral Na + -dependent HCO 3 - transporter NBCn1-mediated HCO 3 - influx in rat medullary thick ascending limb. J Physiol 555: 205-218, 2004.

Orlowski J and Grinstein S. Na + /H + exchangers of mammalian cells. J Biol Chem 272: 22373-22376, 1997.

Ortiz P, Stoos BA, Hong NJ, Boesch DM, Plato CF, and Garvin JL. High-salt diet increases sensitivity to NO and eNOS expression but not NO production in THALs. Hypertension 41: 682-687, 2003.

Ortiz PA. cAMP increases surface expression of NKCC2 in rat thick ascending limbs: role of VAMP. Am J Physiol Renal Physiol 290: F608-F616, 2006.

Ortiz PA and Garvin JL. Autocrine effects of nitric oxide on HCO 3 - transport by rat thick ascending limb. Kidney Int 58: 2069-2074, 2000.

Ortiz PA and Garvin JL. NO inhibits NaCl absorption by rat thick ascending limb through activation of cGMP-stimulated phosphodiesterase. Hypertension 37: 467-471, 2001.

Ortiz PA and Garvin JL. Role of nitric oxide in the regulation of nephron transport. Am J Physiol Renal Physiol 282: F777-F784, 2002.

Ortiz PA, Hong NJ, and Garvin JL. NO decreases thick ascending limb chloride absorption by reducing Na + -K + -2Cl - cotransporter activity. Am J Physiol Renal Physiol 281: F819-F825, 2001.

Ortiz PA, Hong NJ, and Garvin JL. Luminal flow induces eNOS activation and translocation in the rat thick ascending limb. Am J Physiol Renal Physiol 287: F274-F280, 2004.

Ortiz PA, Hong NJ, Plato CF, Varela M, and Garvin JL. An in vivo method for adenovirus-mediated transduction of thick ascending limbs. Kidney Int 63: 1141-1149, 2003.

Plato CF and Garvin JL. 2 -Adrenergic-mediated tubular NO production inhibits thick ascending limb chloride absorption. Am J Physiol Renal Physiol 281: F679-F686, 2001.

Plato CF, Pollock DM, and Garvin JL. Endothelin inhibits thick ascending limb chloride flux via ET B receptor-mediated NO release. Am J Physiol Renal Physiol 279: F326-F333, 2000.

Plato CF, Shesely EG, and Garvin JL. eNOS mediates L -arginine-induced inhibition of thick ascending limb chloride flux. Hypertension 35: 319-323, 2000.

Plato CF, Stoos BA, Wang D, and Garvin JL. Endogenous nitric oxide inhibits chloride transport in the thick ascending limb. Am J Physiol Renal Physiol 276: F159-F163, 1999.

Quentin F, Eladari D, Frische S, Cambillau M, Nielsen S, Alper SL, Paillard M, and Chambrey R. Regulation of the Cl - /HCO 3 - exchanger AE2 in rat thick ascending limb of Henle's loop in response to changes in acid-base and sodium balance. J Am Soc Nephrol 15: 2988-2997, 2004.

Raij L and Baylis C. Glomerular actions of nitric oxide. Kidney Int 48: 20-32, 1995.

Rizzo V, McIntosh DP, Oh P, and Schnitzer JE. In situ flow activates endothelial nitric oxide synthase in luminal caveolae of endothelium with rapid caveolin dissociation and calmodulin association. J Biol Chem 273: 34724-34729, 1998.

Sayago CM and Beierwaltes WH. Nitric oxide synthase and cGMP-mediated stimulation of renin secretion. Am J Physiol Regul Integr Comp Physiol 281: R1146-R1151, 2001.

Schmidt-Nielsen B. The renal concentrating mechanism in insects and mammals: a new hypothesis involving hydrostatic pressures. Am J Physiol Regul Integr Comp Physiol 268: R1087-R1100, 1995.

Shin SJ, Lai FJ, Wen JD, Lin SR, Hsieh MC, Hsiao PJ, and Tsai JH. Increased nitric oxide synthase mRNA expression in the renal medulla of water-deprived rats. Kidney Int 56: 2191-2202, 1999.

Silva G, Beierwaltes WH, and Garvin JL. Extracellular ATP stimulates NO production in rat thick ascending limb. Hypertension 47: 563-567, 2006.

Snyder SH and Bredt DS. Nitric oxide as a neuronal messenger. Trends Pharmacol Sci 12: 125-128, 1991.

Sonntag M, Deussen A, and Schrader J. Role of nitric oxide in local blood flow control in the anaesthetized dog. Pflügers Arch 420: 194-199, 1992.

Turban S, Wang XY, and Knepper MA. Regulation of NHE3, NKCC2, and NCC abundance in kidney during aldosterone escape phenomenon: role of NO. Am J Physiol Renal Physiol 285: F843-F851, 2003.

Ujiie K, Yuen J, Hogarth L, Danziger R, and Star RA. Localization and regulation of endothelial NO synthase mRNA expression in rat kidney. Am J Physiol Renal Fluid Electrolyte Physiol 267: F296-F302, 1994.

Varela M, Herrera M, and Garvin JL. Inhibition of Na-K-ATPase in thick ascending limbs by NO depends on O 2 - and is diminished by a high-salt diet. Am J Physiol Renal Physiol 287: F269-F275, 2004.

Wagner C and Kurtz A. Regulation of renal renin release. Curr Opin Nephrol Hypertens 7: 437-441, 1998.

Watts BA III and Good DW. An apical K + -dependent HCO 3 - transport pathway opposes transepithelial HCO 3 - absorption in rat medullary thick ascending limb. Am J Physiol Renal Physiol 287: F57-F63, 2004.

Welch WJ, Baumgartl H, Lubbers D, and Wilcox CS. Nephron P O 2 and renal oxygen usage in the hypertensive rat kidney. Kidney Int 59: 230-237, 2001.

Welch WJ, Baumgartl H, Lubbers D, and Wilcox CS. Renal oxygenation defects in the spontaneously hypertensive rat: role of AT 1 receptors. Kidney Int 63: 202-208, 2003.

Xan HJ, Kim DH, and Park SH. Regulatory mechanisms of Na/P i cotransporter by glucocorticoid in renal proximal tubule cells: involvement of cAMP and PKC. Kidney Blood Press Res 23: 1-9, 2000.

Yun JC, Oriji G, Gill JR Jr, Coleman BR, Peters J, and Keiser H. Role of muscarinic receptors in renal response to acetylcholine. Am J Physiol Renal Fluid Electrolyte Physiol 265: F46-F52, 1993.


作者单位:1 Hypertension and Vascular Research Division, Henry Ford Hospital, and 2 Department of Physiology, Wayne State University, Detroit, Michigan

作者: Marcela Herrera, Pablo A. Ortiz, and Jeffrey L. Ga 2008-7-4
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具