18 research outputs found

    The functional role of extracellular nucleotides in the renal tubule

    Get PDF
    There is increasing evidence that extracellular nucleotides (such as ATP, ADP, UTP and UDP), as well as the nucleoside adenosine, behave as autocrine or paracrine agents in most tissues including the kidney, acting on a group of receptors known as purinoceptors. Previous studies have shown that activation of these receptors by exogenous nucleotides can influence a variety of renal vascular and tubular functions. Purinoceptors of various subtypes are present on basolateral and apical membranes of renal tubules. However, the extent to which apical receptors are stimulated by endogenous nucleotides is unknown. Using micropuncture, the first part of this study quantified endogenous ATP in the lumen of proximal and distal tubules of the rat in vivo, both under control conditions and during pathophysiological manoeuvres. The results showed that ATP levels were sufficiently high to activate some purinoceptor subtypes. To assess whether the intraluminal ATP was being secreted or merely filtered at the glomerulus, the ATP content of fluid from Bowman's space (in Munich-Wistar rats) was compared with that in proximal tubules. The conclusion was that tubular epithelial cells secrete ATP. Using a proximal tubular epithelial cell line, the mechanism of ATP release was examined. Intracellular stores of ATP were visualised using a marker compound (quinacrine), and the fate of these stores was monitored following hypotonic stimulation of ATP release. The findings suggested that ATP is stored within the cytoplasm, possibly in vesicles, and is released by exocytosis. In the final part of the investigation, using immunohistochemistry, the distribution of five nucleotide-hydrolysing ectonucleotidases, namely NTPDases 1-3, NPP3 and ecto-5'- nucleotidase, was examined along the rat nephron. These enzymes (which differ in their hydrolysis pathways) were found to be differentially expressed along the major segments of the nephron, suggesting that they may be strategically located to influence the activity of the different purinoceptor subtypes

    Pharmacological Properties and Physiological Function of a P2X-Like Current in Single Proximal Tubule Cells Isolated from Frog Kidney

    Get PDF
    Although previous studies have provided evidence for the expression of P2X receptors in renal proximal tubule, only one cell line study has provided functional evidence. The current study investigated the pharmacological properties and physiological role of native P2X-like currents in single frog proximal tubule cells using the whole-cell patch-clamp technique. Extracellular ATP activated a cation conductance (P2Xf) that was also Ca2+-permeable. The agonist sequence for activation was ATP = αβ-MeATP > BzATP = 2-MeSATP, and P2Xf was inhibited by suramin, PPADS and TNP-ATP. Activation of P2Xf attenuated the rundown of a quinidine-sensitive K+ conductance, suggesting that P2Xf plays a role in K+ channel regulation. In addition, ATP/ADP apyrase and inhibitors of P2Xf inhibited regulatory volume decrease (RVD). These data are consistent with the presence of a P2X receptor that plays a role in the regulation of cell volume and K+ channels in frog renal proximal tubule cells

    Ectonucleotidases in the kidney

    No full text
    Members of all four families of ectonucleotidases, namely ectonucleoside triphosphate diphosphohydrolases (NTPDases), ectonucleotide pyrophosphatase/phosphodiesterases (NPPs), ecto-5′-nucleotidase and alkaline phosphatases, have been identified in the renal vasculature and/or tubular structures. In rats and mice, NTPDase1, which hydrolyses ATP through to AMP, is prominent throughout most of the renal vasculature and is also present in the thin ascending limb of Henle and medullary collecting duct. NTPDase2 and NTPDase3, which both prefer ATP over ADP as a substrate, are found in most nephron segments beyond the proximal tubule. NPPs catalyse not only the hydrolysis of ATP and ADP, but also of diadenosine polyphosphates. NPP1 has been identified in proximal and distal tubules of the mouse, while NPP3 is expressed in the rat glomerulus and pars recta, but not in more distal segments. Ecto-5′-nucleotidase, which catalyses the conversion of AMP to adenosine, is found in apical membranes of rat proximal convoluted tubule and intercalated cells of the distal nephron, as well as in the peritubular space. Finally, an alkaline phosphatase, which can theoretically catalyse the entire hydrolysis chain from nucleoside triphosphate to nucleoside, has been identified in apical membranes of rat proximal tubules; however, this enzyme exhibits relatively high Km values for adenine nucleotides. Although information on renal ectonucleotidases is still incomplete, the enzymes’ varied distribution in the vasculature and along the nephron suggests that they can profoundly influence purinoceptor activity through the hydrolysis, and generation, of agonists of the various purinoceptor subtypes. This review provides an update on renal ectonucleotidases and speculates on the functional significance of these enzymes in terms of glomerular and tubular physiology and pathophysiology

    A critical function for CD200 in lung immune homeostasis and the severity of influenza infection.

    No full text
    The lung must maintain a high threshold of immune 'ignorance' to innocuous antigens to avoid inflammatory disease that depends on the balance of positive inflammatory signals and repressor pathways. We demonstrate here that airway macrophages had higher expression of the negative regulator CD200 receptor (CD200R) than did their systemic counterparts. Lung macrophages were restrained by CD200 expressed on airway epithelium. Mice lacking CD200 had more macrophage activity and enhanced sensitivity to influenza infection, which led to delayed resolution of inflammation and, ultimately, death. The administration of agonists that bind CD200R, however, prevented inflammatory lung disease. Thus, CD200R is critical for lung macrophage immune homeostasis in the resting state and limits inflammatory amplitude and duration during pulmonary influenza infection

    P2 receptors in renal pathophysiology

    No full text
    Our knowledge and understanding of the P2 receptor signalling system in the kidney have increased significantly in the last ten years. The broad range of physiological roles proposed for this receptor system and the variety of P2 receptor subtypes found in the kidney suggest that any disturbance of function may contribute to several pathological processes. So far, most reports of a possible pathophysiological role for this system in the kidney have focussed on polycystic kidney disease, where abnormal P2 receptor signalling might be involved in cyst expansion and disease progression, and on the P2X7 receptor, a unique P2X subtype, which when activated enhances inflammatory cytokine release and production, and also cell death. Expression of this particular receptor is upregulated in some forms of chronic renal injury and inflammatory diseases. Further studies of adenosine triphosphate signalling and P2 receptor expression in renal disorders could provide us with novel insights into the role of these receptors in both normal and abnormal kidney function

    Effects of extracellular nucleotides on renal tubular solute transport

    Get PDF
    A range of P2 receptor subtypes has been identified along the renal tubule, in both apical and basolateral membranes. Furthermore, it has been shown that nucleotides are released from renal tubular cells, and that ectonucleotidases are present in several nephron segments. These findings suggest an autocrine/paracrine role for nucleotides in regulating tubular function. The present review catalogues the known actions of extracellular nucleotides on tubular solute transport. In the proximal tubule, there is firm evidence that stimulation of apical P2Y1 receptors inhibits bicarbonate reabsorption, whilst basolaterally applied ATP has the opposite effect. Clearance studies suggest that systemic diadenosine polyphosphates profoundly reduce proximal tubular fluid transport, through as yet unidentified P2 receptors. To date, only circumstantial evidence is available for an action of nucleotides on transport in the loop of Henle; and no studies have been made on native distal tubules, though observations in cell lines suggest an inhibitory effect on sodium, calcium and magnesium transport. The nephron segment most studied is the collecting duct. Apically applied nucleotides inhibit the activity of small-conductance K+ channels in mouse collecting duct, apparently through stimulation of P2Y2 receptors. There is also evidence, from cell lines and native tissue, that apically (and in some cases basolaterally) applied nucleotides inhibit sodium reabsorption. In mice pharmacological profiling implicates P2Y2 receptors; but in rats, the receptor subtype(s) responsible is/are unclear. Recent patch-clamp studies in rat collecting ducts implicate apical P2Y and P2X subtypes, with evidence for both inhibitory and stimulatory effects. Despite considerable progress, clarification of the physiological role of the tubular P2 receptor system remains some way off

    ENaC, renal sodium excretion and extracellular ATP

    No full text
    Sodium balance determines the extracellular fluid volume and sets arterial blood pressure (BP). Chronically raised BP (hypertension) represents a major health risk in Western societies. The relationship between BP and renal sodium excretion (the pressure/natriuresis relationship) represents the key element in defining the BP homeostatic set point. The renin–angiotensin–aldosterone system (RAAS) makes major adjustments to the rates of renal sodium secretion, but this system works slowly over a period of hours to days. More rapid adjustments can be made by the sympathetic nervous system, although the kidney can function well without sympathetic nerves. Attention has now focussed on regulatory mechanisms within the kidney, including extracellular nucleotides and the P2 receptor system. Here, we discuss how extracellular ATP can control renal sodium excretion by altering the activity of epithelial sodium channels (ENaC) present in the apical membrane of principal cells. There remains considerable controversy over the molecular targets for released ATP, although the P2Y2 receptor has received much attention. We review the available data and reflect on our own findings in which ATP-activated P2Y and P2X receptors make adjustments to ENaC activity and therefore sodium excretion
    corecore