Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Physiol Res ; 69(3): 353-369, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32301620

ABSTRACT

Proper renal blood flow (RBF) and glomerular filtration rate (GFR) are critical for maintaining normal blood pressure, kidney function and water and electrolyte homeostasis. The renal microvasculature expresses a multitude of receptors mediating vasodilation and vasoconstriction, which can influence glomerular blood flow and capillary pressure. Despite this, RBF and GFR remain quite stable when arterial pressure fluctuates because of the autoregulatory mechanism. ATP and adenosine participate in autoregulatory control of RBF and GFR via activation of two different purinoceptor families (P1 and P2). Purinoceptors are widely expressed in renal microvasculature and tubules. Emerging data show altered purinoceptor signaling in hypertension-associated kidney injury, diabetic nephropathy, sepsis, ischemia-reperfusion induced acute kidney injury and polycystic kidney disease. In this brief review, we highlight recent studies and new insights on purinoceptors regulating renal microvascular function and renal hemodynamics. We also address the mechanisms underlying renal microvascular injury and impaired renal autoregulation, focusing on purinoceptor signaling and hypertension-induced renal microvascular dysfunction. Interested readers are directed to several excellent and comprehensive reviews that recently covered the topics of renal autoregulation, and nucleotides in kidney function under physiological and pathophysiological conditions (Inscho 2009, Navar et al. 2008, Carlstrom et al. 2015, Vallon et al. 2020).


Subject(s)
Hypertension/physiopathology , Kidney/blood supply , Kidney/physiopathology , Receptors, Purinergic/metabolism , Animals , Glomerular Filtration Rate , Homeostasis , Humans , Hypertension/metabolism , Kidney/metabolism , Renal Circulation/physiology
2.
Acta Physiol (Oxf) ; 222(2)2018 02.
Article in English | MEDLINE | ID: mdl-28640982

ABSTRACT

AIM: Sphingosine-1-phosphate (S1P) influences resistance vessel function and is implicated in renal pathological processes. Previous studies revealed that S1P evoked potent vasoconstriction of the pre-glomerular microvasculature, but the underlying mechanisms remain incompletely defined. We postulated that S1P-mediated pre-glomerular microvascular vasoconstriction involves activation of voltage-dependent L-type calcium channels (L-VDCC) and the rho/rho kinase pathway. METHODS: Afferent arteriolar reactivity was assessed in vitro using the blood-perfused rat juxtamedullary nephron preparation, and diameter was measured during exposure to physiological and pharmacological agents. RESULTS: Exogenous S1P (10-9 -10-5 mol L-1 ) evoked concentration-dependent vasoconstriction of afferent arterioles. Superfusion with nifedipine, a L-VDCC blocker, increased arteriolar diameter by 39 ± 18% of baseline and significantly attenuated the S1P-induced vasoconstriction. Superfusion with the rho kinase inhibitor, Y-27632, increased diameter by 60 ± 12% of baseline and also significantly blunted vasoconstriction by S1P. Combined nifedipine and Y-27632 treatment significantly inhibited S1P-induced vasoconstriction over the entire concentration range tested. In contrast, depletion of intracellular Ca2+ stores with the Ca2+ -ATPase inhibitors, thapsigargin or cyclopiazonic acid, did not alter the S1P-mediated vasoconstrictor profile. Scavenging reactive oxygen species (ROS) or inhibition of nicotinamide adenine dinucleotide phosphate oxidase activity significantly attenuated S1P-mediated vasoconstriction. CONCLUSION: Exogenous S1P elicits potent vasoconstriction of rat afferent arterioles. These data also demonstrate that S1P-mediated pre-glomerular vasoconstriction involves activation of L-VDCC, the rho/rho kinase pathway and ROS. Mobilization of Ca2+ from intracellular stores is not required for S1P-mediated vasoconstriction. These studies reveal a potential role for S1P in the modulation of renal microvascular tone.


Subject(s)
Arterioles/metabolism , Kidney/blood supply , Kidney/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Vasoconstriction/physiology , Animals , Arterioles/drug effects , Lysophospholipids/pharmacology , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Renal Circulation/physiology , Sphingosine/metabolism , Sphingosine/pharmacology , Vasoconstriction/drug effects
3.
Acta Physiol (Oxf) ; 221(3): 204-220, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28544543

ABSTRACT

AIM: Little is known about how toll-like receptor 4 (TLR4) influences the renal microvasculature. We hypothesized that acute TLR4 stimulation with lipopolysaccharide (LPS) impairs afferent arteriole autoregulatory behaviour, partially through reactive oxygen species (ROS). METHODS: We assessed afferent arteriole autoregulatory behaviour after LPS treatment (1 mg kg-1 ; i.p.) using the in vitro blood-perfused juxtamedullary nephron preparation. Autoregulatory behaviour was assessed by measuring diameter responses to stepwise changes in renal perfusion pressure. TLR4 expression was assessed by immunofluorescence, immunohistochemistry and Western blot analysis in the renal cortex and vasculature. RESULTS: Baseline arteriole diameter at 100 mmHg averaged 15.2 ± 1.2 µm and 12.2 ± 1.0 µm for control and LPS groups (P < 0.05) respectively. When perfusion pressure was increased in 15 mmHg increments from 65 to 170 mmHg, arteriole diameter in control kidneys decreased significantly to 69 ± 6% of baseline diameter. In the LPS-treated group, arteriole diameter remained essentially unchanged (103 ± 9% of baseline), indicating impaired autoregulatory behaviour. Pre-treatment with anti-TLR4 antibody or the TLR4 antagonist, LPS-RS, preserved autoregulatory behaviour during LPS treatment. P2 receptor reactivity was normal in control and LPS-treated rats. Pre-treatment with Losartan (angiotensin type 1 receptor blocker; (AT1 ) 2 mg kg-1 ; i.p.) increased baseline afferent arteriole diameter but did not preserve autoregulatory behaviour in LPS-treated rats. Acute exposure to Tempol (10-3 mol L-1 ), a superoxide dismutase mimetic, restored pressure-mediated vasoconstriction in kidneys from LPS-treated rats. CONCLUSION: These data demonstrate that TLR4 activation impairs afferent arteriole autoregulatory behaviour, partially through ROS, but independently of P2 and AT1 receptor activation.


Subject(s)
Kidney/blood supply , Toll-Like Receptor 4/metabolism , Vasodilation/drug effects , Vasodilation/physiology , Animals , Antibodies , Arterioles/drug effects , Arterioles/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Lipopolysaccharides/toxicity , Male , Rats , Reactive Oxygen Species , Toll-Like Receptor 4/genetics
4.
Acta Physiol Hung ; 96(3): 375-82, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19706379

ABSTRACT

This article pays tribute to the tremendous achievements of Dr. László Rosivall in renal (patho)physiology research and nephrology education in Hungary on the occasion of his 60th birthday. For the past several decades Dr. Rosivall has been a charismatic leader of academic institutions, national and international societies, foundations in physiology, nephrology and hypertension, but the most important of his many contributions, is his role as a scientist. He earned his MD with Summa cum Laude at Semmelweis University (1973) and was invited immediately after that to join the laboratory of Hársing. He studied the distribution of intra-renal blood flow employing then state-of-the-art methods as well as developed his own technique at Semmelweis University and at the University of Bergen with Knut Aukland. This led to his PhD thesis and degree in 1980. An important determinant of his early basic scientific training and development was his postdoctoral research fellowship and later many visiting professorships in the Nephrology Research and Training Center (NRTC) at the University of Alabama at Birmingham, Birmingham, AL, USA between 1981 and 1983. Actually, this research fellowship not only impacted his own future career, but it also cleared the path for many other young Hungarian scientists who later trained with Dr. Rosivall and then at UAB. The early 1980s were the years of significant scientific discoveries and the NRTC team at UAB made important contributions by their studies on renal and glomerular hemodynamics, the renin-angiotensin system (12, 19, 22) and by the development of classic experimental techniques like renal micropuncture, microperfusion, and the juxtamedullary nephron preparation (3) that are still being used worldwide. When Dr. Rosivall joined UAB in the 1980s, the team at the NRTC included Drs. Navar, Bell, Inscho, Carmines, Casellas, and Oparil, among many others, who share their fond memories of working with Dr. Rosivall in this article.


Subject(s)
Biomedical Research/history , Nephrology/history , History, 20th Century , Hungary , Nephrology/education
5.
Acta Physiol Scand ; 181(4): 445-53, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15283757

ABSTRACT

Autoregulation of renal blood flow is an established physiological phenomenon, however the signalling mechanisms involved remain elusive. Autoregulatory adjustments in preglomerular resistance involve myogenic and tubuloglomerular feedback (TGF) influences. While there is general agreement on the participation of these two regulatory pathways, the signalling molecules and effector mechanisms have not been identified. Currently, there are two major hypotheses being considered to explain the mechanism by which TGF signals are transmitted from the macula densa to the afferent arteriole. The adenosine hypothesis proposes that the released adenosine triphosphate (ATP) is hydrolysed to adenosine and this product stimulates preglomerular vasoconstriction by activation of A(1) receptors on the afferent arteriole. Alternatively, the P2 receptor hypothesis postulates that ATP released from the macula densa directly stimulates afferent arteriolar vasoconstriction by activation of ATP-sensitive P2X(1) receptors. This hypothesis has emerged from the realization that P2X(1) receptors are heavily expressed along the preglomerular vasculature. Inactivation of P2X(1) receptors impairs autoregulatory responses while afferent arteriolar responses to A(1) adenosine receptor activation are retained. Autoregulatory behaviour is markedly attenuated in mice lacking P2X(1) receptors but responses to adenosine A(1) receptor activation remain intact. More recent experiments suggest that P2X(1) receptors play an essential role in TGF-dependent vasoconstriction of the afferent arteriole. Interruption of TGF-dependent influences on afferent arteriolar diameter, by papillectomy or furosemide treatment, significantly attenuated pressure-mediated afferent arteriolar vasoconstriction in wild-type mice but had no effect on the response in P2X(1) knockout mice. Collectively, these observations support an essential role for P2X(1) receptors in TGF-mediated afferent arteriolar vasoconstriction.


Subject(s)
Receptors, Purinergic P2/physiology , Renal Circulation/physiology , Adenosine Triphosphate/physiology , Animals , Arterioles/physiology , Homeostasis/physiology , Mice , Mice, Knockout , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X , Transforming Growth Factors/physiology
6.
Am J Physiol Heart Circ Physiol ; 281(5): H2089-96, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11668070

ABSTRACT

This study was conducted to test the hypothesis that the cytochrome P-450 (CYP450) metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) contributes to the afferent arteriolar response to P2 receptor activation. Afferent arteriolar responses to ATP, the P2X agonist, alpha,beta-methylene ATP and the P2Y agonist UTP were determined before and after treatment with the selective CYP450 hydroxylase inhibitor, N-methylsulfonyl-12,12-dibromododec-11-enamide (DDMS) or the 20-HETE antagonist, 20-hydroxyeicosa-6(Z),15(Z)-dienoic acid (20-HEDE). Stimulation with 1.0 and 10 microM ATP elicited an initial preglomerular vasoconstriction of 12 +/- 1% and 45 +/- 4% and a sustained vasoconstriction of 11 +/- 1% and 11 +/- 2%, respectively. DDMS or 20-HEDE significantly attenuated the sustained afferent arteriolar constrictor response to ATP. alpha,beta-Methylene ATP (1 microM) induced a rapid initial afferent vasoconstriction of 64 +/- 3%, which partially recovered to a stable diameter 10 +/- 1% smaller than control. Both DDMS and 20-HEDE significantly attenuated the initial vasoconstriction and abolished the sustained vasoconstrictor response to alpha,beta-methylene ATP. UTP decreased afferent diameter by 50 +/- 5% and 20-HEDE did not change this response. In addition, the ATP-induced increase in the intracellular Ca2+ concentration in preglomerular microvascular smooth muscle cells was significantly attenuated by 20-HEDE. Taken together, these results are consistent with the hypothesis that the CYP450 metabolite 20-HETE participates in the afferent arteriolar response to activation of P2X receptors.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Receptors, Purinergic P2/metabolism , Vasoconstriction/physiology , Adenosine Triphosphate/pharmacology , Amides/pharmacology , Animals , Arterioles/physiology , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hydroxyeicosatetraenoic Acids/antagonists & inhibitors , Male , Muscle, Smooth, Vascular/physiology , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X , Receptors, Purinergic P2Y1 , Renal Artery/physiology , Sulfones/pharmacology , Uridine Triphosphate/metabolism , Vasoconstriction/drug effects
7.
Clin Exp Pharmacol Physiol ; 28(4): 332-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11339210

ABSTRACT

1. The field of extracellular nucleotides and purinoceptors has undergone a resurgence of interest and enthusiasm in the past decade. More and more investigators are probing the physiological and pathophysiological roles of P2 receptors in virtually every organ system, including the kidney. 2. With this renewed interest has come a new appreciation for the roles extracellular adenine nucleotides can play in regulating or modulating renal function. In the past 5 years, investigators have provided compelling evidence that extracellular nucleotides, working through activation of P2 purinoceptors, have a significant impact on renal microvascular function, mesangial cell function and on renal epithelial transport. 3. Evidence has been uncovered that implicates P2 receptor activation in mediating renal microvascular autoregulatory behaviour. Locally released ATP has a direct paracrine and/or autocrine effect modulating renal epithelial transporters and tubular epithelial channels to influence tubular fluid composition. 4. While the specific roles of extracellular nucleotides and their receptors in the kidney have not been absolutely identified, it now appears clear that endogenously released ATP may play a significant role in regulating kidney function. 5. The purpose of the present review is to update our current understanding of the effect of P2 receptor activation on renal microvascular function and to detail the signal transduction mechanisms known to be involved.


Subject(s)
Adenosine Triphosphate/pharmacology , Calcium Signaling/drug effects , Kidney/drug effects , Receptors, Purinergic P2/drug effects , Vasoconstriction/drug effects , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Calcium Signaling/physiology , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Humans , Kidney/blood supply , Kidney/physiology , Kidney Glomerulus/drug effects , Kidney Glomerulus/physiology , Microcirculation , Platelet Aggregation Inhibitors/pharmacology , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Receptors, Purinergic P2/physiology , Renal Artery/drug effects , Renal Artery/physiology , Vasoconstriction/physiology
8.
Am J Physiol Renal Physiol ; 280(6): F927-44, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352833

ABSTRACT

In the last 10-15 years, interest in the physiological role of P2 receptors has grown rapidly. Cellular, tissue, and organ responses to P2 receptor activation have been described in numerous in vivo and in vitro models. The purpose of this review is to provide an update of the recent advances made in determining the involvement of P2 receptors in the control of renal hemodynamics and the renal microcirculation. Special attention will be paid to work published in the last 5-6 years directed at understanding the role of P2 receptors in the physiological control of renal microvascular function. Several investigators have begun to evaluate the effects of P2 receptor activation on renal microvascular function across several species. In vivo and in vitro evidence consistently supports the hypothesis that P2 receptor activation by locally released extracellular nucleotides influences microvascular function. Extracellular nucleotides selectively influence preglomerular resistance without having an effect on postglomerular tone. P2 receptor inactivation blocks autoregulatory behavior whereas responsiveness to other vasoconstrictor agonists is retained. P2 receptor stimulation activates multiple intracellular signal transduction pathways in preglomerular smooth muscle cells and mesangial cells. Renal microvascular cells and mesangial cells express multiple subtypes of P2 receptors; however, the specific role each plays in regulating vascular and mesangial cell function remains unclear. Accordingly, the results of studies performed to date provide strong support for the hypothesis that P2 receptors are important contributors to the physiological regulation of renal microvascular and/or glomerular function.


Subject(s)
Receptors, Purinergic P2/physiology , Renal Circulation/physiology , Animals , Humans , Microcirculation/physiology
9.
Am J Physiol Renal Physiol ; 280(6): F1054-61, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352845

ABSTRACT

This study tested the hypothesis that P2X receptor activation increases intracellular Ca(2+) concentration ([Ca(2+)](i)) in preglomerular microvascular smooth muscle cells (MVSMC) by evoking voltage-dependent calcium influx. MVSMC were obtained and loaded with the calcium-sensitive dye fura 2 and studied by using single-cell fluorescence microscopy. The effect of P2X receptor activation on [Ca(2+)](i) was assessed by using the P2X receptor-selective agonist alpha,beta-methylene-ATP and was compared with responses elicited by the endogenous P2 receptor agonist ATP. alpha,beta-Methylene-ATP increased [Ca(2+)](i) dose dependently. Peak increases in [Ca(2+)](i) averaged 37 +/- 11, 73 +/- 15, and 103 +/- 21 nM at agonist concentrations of 0.1, 1, and 10 microM, respectively. The average peak response elicited by 10 microM alpha,beta-methylene-ATP was approximately 34% of the response obtained with 10 microM ATP. alpha,beta-Methylene-ATP induced a transient increase in [Ca(2+)](i) before [Ca(2+)](i) returned to baseline, whereas ATP induced a biphasic response including a peak response followed by a sustained plateau. In Ca(2+)-free medium, ATP induced a sharp transient increase in [Ca(2+)](i), whereas the response to alpha,beta-methylene-ATP was abolished. Ca(2+) channel blockade with 10 microM diltiazem or nifedipine attenuated the response to alpha,beta-methylene-ATP, whereas nonspecific blockade of Ca(2+) influx pathways with 5 mM Ni(2+) abolished the response. Blockade of P2X receptors with the novel P2X receptor antagonist NF-279 completely but reversibly abolished the response to alpha,beta-methylene-ATP. These results indicate that P2X receptor activation by alpha,beta-methylene-ATP increases [Ca(2+)](i) in preglomerular MVSMC, in part, by stimulating voltage-dependent Ca(2+) influx through L-type Ca(2+) channels.


Subject(s)
Calcium Signaling/physiology , Kidney Glomerulus/blood supply , Muscle, Smooth, Vascular/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Diltiazem/pharmacology , In Vitro Techniques , Male , Nickel/pharmacology , Rats , Rats, Sprague-Dawley
10.
Am J Physiol Renal Physiol ; 280(3): F406-14, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181402

ABSTRACT

Adenosine vasoconstricts preglomerular arterioles via adenosine A1 receptors. Because adenosine also activates adenosine A2 receptors, its overall renal vascular actions are complex and not fully understood. The present study was performed to determine the relative contributions of adenosine A1 and A2a receptors to the responsiveness of the renal microvasculature to adenosine. Afferent and efferent arteriolar diameters were monitored in vitro using the blood-perfused rat juxtamedullary nephron preparation. Basal afferent and efferent arteriolar diameters averaged 17.1 +/- 0.5 (n = 35) and 17.8 +/- 0.5 (n = 20) microm, respectively. Superfusion with 0.1 and 1 micromol/l adenosine did not significantly alter afferent and efferent arteriolar diameters; however, 10 micromol/l adenosine significantly reduced afferent and efferent arteriolar diameters (-8.2 +/- 0.8 and -5.7 +/- 0.6%, respectively). The afferent and efferent arteriolar vasoconstrictor responses to adenosine waned at a dose of 100 micromol/l, such that diameters returned to values not significantly different from control within 2 min. During adenosine A1 receptor blockade with 8-noradamantan-3-yl-1,3-dipropylxanthine (KW-3902: 10 micromol/l), 10 and 100 micromol/l adenosine significantly increased afferent diameter by, respectively, 8.1 +/- 1.2 and 13.7 +/- 1.3% (n = 14) and efferent arteriolar diameter by 6.4 +/- 1.3 and 9.3 +/- 1.2% (n = 8). The afferent and efferent arteriolar vasodilatory responses to adenosine in the presence of KW-3902 were significantly attenuated by addition of the adenosine A2a receptor antagonist 1,3-dipropyl-7-methyl-8-(3,4-dimethoxystyryl)xanthine (KF-17837: 15 micromol/l, n = 7 and 6, respectively). The addition of KF-17837 alone significantly enhanced afferent (n = 15) and efferent (n = 6) arteriolar vasoconstrictor responses to 1, 10, and 100 micromol/l adenosine. These results indicate the presence of adenosine A1 and A2a receptors on afferent and efferent arterioles of juxtamedullary nephrons, such that adenosine A2a receptor-mediated vasodilation partially buffers adenosine-induced vasoconstriction in both pre- and postglomerular segments of the renal microvasculature.


Subject(s)
Receptors, Purinergic P1/physiology , Renal Circulation/physiology , Adenosine/metabolism , Adenosine/pharmacology , Animals , Arterioles/drug effects , Arterioles/physiology , Extracellular Space/metabolism , Male , Microcirculation/physiology , Purinergic P1 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A , Vasomotor System/physiology , Xanthines/pharmacology
11.
Methods Mol Med ; 51: 435-49, 2001.
Article in English | MEDLINE | ID: mdl-21331735

ABSTRACT

Understanding the endocrine, paracrine, and autocrine mechanisms involved in the regulation of renal hemodynamics has proven an elusive quest (1). What has emerged, however, is a greater appreciation for the elegance and complexity of the renal microcirculation, which provides exquisite control of renal vascular resistance.

12.
Hypertension ; 35(1 Pt 2): 280-6, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10642311

ABSTRACT

This study was performed to test the hypothesis that endothelin peptides differentially influence intracellular calcium concentration ([Ca(2+)](i)) in preglomerular microvascular smooth muscle cells (MVSMC), in part through activation of endothelin (ET)(A) receptors. Experiments were performed in vitro with the use of single MVSMC freshly isolated from rat preglomerular microvessels. The effect of ET-1, ET-2, and ET-3 on [Ca(2+)](i) was measured with the use of the calcium-sensitive dye, fura 2, and standard fluorescence microscopy techniques. Baseline [Ca(2+)](i) averaged 84+/-3 nmol/L (n=141 cells from 23 dispersions). ET-1 concentrations of 1, 10, and 100 nmol/L evoked peak increases in [Ca(2+)](i) of 48+/-16, 930+/-125, and 810+/-130 nmol/L, respectively. The time course of the [Ca(2+)](i) response was biphasic, beginning with a rapid initial increase followed by a sustained plateau phase or a period during which [Ca(2+)](i) oscillated sharply. Similar responses were observed after ET-2 administration. In contrast, ET-3 stimulated monophasic increases in [Ca(2+)](i) of only 14+/-5, 33+/-16, and 44+/-19 nmol/L at peptide concentrations of 1, 10, and 100 nmol/L, respectively. These responses are significantly smaller than responses to ET-1 or ET-2, respectively. The relative contributions of calcium mobilization and calcium influx in the response to ET-1 were also evaluated. Removal of calcium from the bathing medium did not significantly alter the peak response to 10 nmol/L ET-1 but abolished the late phase elevation of [Ca(2+)](i). These data demonstrate that endothelin peptides increase [Ca(2+)](i) in preglomerular MVSMC. The concentration-response profiles are consistent with the response involving activation of ET(A) receptors. Furthermore, these results suggest that ET-1 increases [Ca(2+)](i) by stimulating both the release of intracellular calcium and the influx of calcium from the extracellular medium.


Subject(s)
Calcium Signaling/drug effects , Endothelin-1/pharmacology , Kidney Glomerulus/blood supply , Muscle, Smooth, Vascular/physiology , Animals , Calcium/pharmacokinetics , Calcium Signaling/physiology , Cells, Cultured , Endothelin-2/pharmacology , Endothelin-3/pharmacology , Extracellular Space/metabolism , Microcirculation/physiology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , Rats , Rats, Sprague-Dawley , Receptors, Endothelin/physiology , Renal Circulation/physiology
13.
Hypertension ; 35(1 Pt 2): 307-12, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10642316

ABSTRACT

Arachidonic acid metabolites contribute to the endothelin-1 (ET-1)-induced decrease in renal blood flow, but the vascular sites of action are unknown. Experiments performed in vitro used the rat juxtamedullary nephron preparation combined with videomicroscopy. The response of afferent arterioles to ET-1 was determined before and after cytochrome P450 (CYP450) or cyclooxygenase (COX) inhibition. Afferent arteriolar diameter averaged 20+/-1 microm (n=17) at a renal perfusion pressure of 100 mm Hg. Superfusion with 0.001 to 10 nmol/L ET-1 caused a graded decrease in diameter of the afferent arteriole. Vessel diameter decreased by 30+/-2% and 41+/-2% in response to 1 and 10 nmol/L ET-1, respectively. The afferent arteriolar response to ET-1 was significantly attenuated during administration of the CYP450 hydroxylase inhibitor N-methylsulfonyl-12,12-dibromododec-11-enamide (DDMS), such that afferent arteriolar diameter decreased by 19+/-3% and 22+/-3% in response to 1 and 10 nmol/L ET-1, respectively. COX inhibition also greatly attenuated the vasoconstriction elicited by ET-1, whereas the CYP450 epoxygenase inhibitor N-methylsulfonyl-6-(2-proparglyoxyphenyl) hexanamide enhanced the ET-1-mediated vascular response. Additional studies were performed using freshly isolated smooth muscle cells prepared from preglomerular microvessels. Renal microvascular smooth muscle cells were loaded with the calcium-sensitive dye fura 2 and studied by use of single-cell fluorescence microscopy. Basal renal microvascular smooth muscle cell [Ca(2+)](i) averaged 95+/-3 nmol/L (n=42). ET-1 (10 nmol/L) increased microvascular smooth muscle cell [Ca(2+)](i) to a peak value of 731+/-75 nmol/L before stabilizing at 136+/-8 nmol/L. Administration of DDMS or the COX inhibitor indomethacin significantly attenuated the renal microvascular smooth muscle cell calcium response to ET-1. These data demonstrate that CYP450 hydroxylase and COX arachidonic acid metabolites contribute importantly to the afferent arteriolar diameter and renal microvascular smooth muscle cell calcium responses elicited by ET-1.


Subject(s)
Calcium/metabolism , Cytochrome P-450 Enzyme System/metabolism , Endothelin-1/pharmacology , Muscle, Smooth, Vascular/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Amides/pharmacology , Animals , Arterioles/drug effects , Arterioles/physiology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cyclooxygenase Inhibitors/pharmacology , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Indomethacin/pharmacology , Male , Rats , Rats, Sprague-Dawley , Renal Artery/cytology , Renal Circulation/drug effects , Renal Circulation/physiology , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Sulfones/pharmacology , Vasoconstriction/drug effects , Vasoconstriction/physiology
14.
Gen Pharmacol ; 34(6): 409-15, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11483290

ABSTRACT

The current study was performed to determine the effect of calcium store depletion with cyclopiazonic acid (CPA) on the pre- and postglomerular vasoconstrictor responses to angiotensin II (ANG II) and norepinephrine (NE). CPA treatment significantly attenuated the afferent arteriolar response to 10 nM ANG II by 51% and to 1000 nM NE by 19%. Efferent arteriolar responses to ANG II and NE were also greatly attenuated in the presence of CPA. These data demonstrate that afferent and efferent arteriolar responses to ANG II and NE depend on release of calcium from CPA-sensitive intracellular stores. Furthermore, the postglomerular response to these agents exhibits a greater dependency on calcium release from intracellular stores.


Subject(s)
Angiotensin II/pharmacology , Calcium/metabolism , Kidney Glomerulus/drug effects , Norepinephrine/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Arterioles/drug effects , Indoles/pharmacology , Kidney Glomerulus/blood supply , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Time Factors
15.
Cell Immunol ; 195(2): 147-56, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10448014

ABSTRACT

While it is recognized that activated dendritic cells perform their immune functions with greater efficacy, it is not altogether clear what factors are responsible for such activation. Recent evidence points to an important role for extracellular nucleotides in the modulation of leukocyte function. In the present study we investigated the ability of extracellular nucleotides to activate CD11c(+) murine dendritic cells. Mobilization of intracellular calcium was observed following treatment of these cells with UTP or UDP, but not ATP. Furthermore, this nucleotide receptor was pertussis toxin-sensitive, suggesting the presence of a P2Y nucleotide receptor. Such receptors were not present on murine peritoneal macrophages or on CD11c-negative leukocyte populations. Importantly, activation of these P2Y nucleotide receptors on dendritic cells provided a potent stimulus for cytokine mRNA expression and secretion. Thus, expression of a P2Y nucleotide receptor on CD11c(+) dendritic cells functions to mobilize intracellular calcium and to induce cytokine production.


Subject(s)
Dendritic Cells/metabolism , Interleukins/biosynthesis , Receptors, Purinergic P2/metabolism , Uracil Nucleotides/pharmacology , Adenine Nucleotides/pharmacology , Animals , Calcium/metabolism , Cell Separation , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Interleukins/genetics , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Pertussis Toxin , Purinergic P2 Receptor Agonists , Purinergic P2 Receptor Antagonists , Suramin/pharmacology , Thapsigargin/pharmacology , Uracil Nucleotides/antagonists & inhibitors , Virulence Factors, Bordetella/pharmacology
16.
Br J Pharmacol ; 127(6): 1399-405, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10455289

ABSTRACT

Previous studies have demonstrated an important role for the cytochrome P450 (CYT-P450) pathway in afferent arteriole autoregulatory responses but the involvement of specific pathways remains unknown. Experiments were performed to determine the role of CYT-P450 epoxygenase and hydroxylase pathways in pressure mediated preglomerular autoregulatory responses. Afferent arteriolar diameter was measured as renal perfusion pressure was increased from 80-160 mmHg. Afferent arteriolar diameter averaged 19+/-2 microm at a renal perfusion pressure of 80 mmHg and decreased by 15+/-2% when pressure was increased to 160 mmHg. Inhibition of the epoxygenase pathway with 6-(2-proparglyloxyphenyl)hexanoic acid (PPOH), enhanced the microvascular response to increasing renal perfusion pressure. In the presence of 50 microM PPOH, afferent arteriolar diameter decreased by 29+/-4% when pressure was increased from 80-160 mmHg. Likewise, the sulphonimide derivative of PPOH, N-methylsulphonyl-6-(2-proparglyloxyphenyl) hexanamide (MS-PPOH, 50 microM), enhanced the afferent arteriolar response to increasing renal perfusion pressure. In contrast, the selective CYT-P450 hydroxylase inhibitor, N-methylsulphonyl-12,12-dibromododec-11-enamide (DDMS) attenuated the vascular response to increasing renal perfusion pressure. In the pressure of 25 microM DDMS, afferent arteriolar diameter decreased by 4+/-2% when pressure was increased from 80-160 mmHg. These results suggest that CYT-P450 metabolites of the epoxygenase pathway alter afferent arteriolar responsiveness and thereby modify the ability of the preglomerular vasculature to autoregulate renal blood flow. Additionally, these results provide further support to the concept that a metabolite of the hydroxylase pathway is an integral component of the afferent arteriolar response to elevations in perfusion pressure.


Subject(s)
Amides/pharmacology , Arterioles/physiology , Caproates/pharmacology , Cytochrome P-450 Enzyme System/physiology , Mixed Function Oxygenases/physiology , Oxygenases/physiology , Animals , Arterioles/drug effects , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Homeostasis , Kidney/blood supply , Male , Mixed Function Oxygenases/antagonists & inhibitors , Oxygenases/antagonists & inhibitors , Perfusion , Pressure , Rats , Rats, Sprague-Dawley , Renal Artery/drug effects , Renal Artery/physiology , Signal Transduction/drug effects , Sulfones/pharmacology
17.
J Am Soc Nephrol ; 10(3): 492-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10073599

ABSTRACT

Recent studies have suggested a role for P2 purinoceptors on vascular smooth muscle cells in the mechanism of renal autoregulation. Experiments were performed in anesthetized dogs (n = 9) to examine renal blood flow (RBF) autoregulatory efficiency before and after saturation of P2 purinoceptors with acute intra-arterial administration of ATP (1 mg/kg per min). Dogs were pretreated with the nitric oxide synthase inhibitor nitro-L-arginine (NLA) (50 microg/kg per min), to avoid endothelial P2 receptor-mediated effects on nitric oxide release caused by the intra-arterial ATP infusions. NLA treatment decreased RBF (5.3+/-0.3 to 3.6+/-0.2 ml/min per g) and sodium excretion (3.6+/-0.4 to 0.9+/-0.2 ml/min per g) without producing significant changes in GFR (0.92+/-0.04 to 0.90+/-0.06 ml/min per g) or RBF autoregulatory efficiency. ATP administration to NLA-treated dogs resulted in further decreases in RBF (2.8+/-0.2 ml/min per g), GFR (0.58+/-0.05 ml/min per g), and sodium excretion (0.6+/-0.2 micromol/min per g). In addition, there was marked impairment of RBF autoregulatory efficiency during ATP infusion. The slopes of the arterial pressure-blood flow relationships at renal arterial pressures of >75 mmHg were significantly altered, from 0.003+/-0.001 to 0.2+/-0.002 ml/min per g per mmHg. Discontinuation of ATP infusion restored RBF autoregulatory efficiency. Norepinephrine (5 microg/kg per min) administration in these NLA-treated dogs decreased RBF (2.5+/-0.3 ml/min per g; n = 4) to a similar extent, compared with ATP, but did not impair RBF autoregulation. These results support the hypothesis that P2 purinoceptors may be involved in mediating autoregulatory adjustments in renal vascular resistance.


Subject(s)
Adenosine Triphosphate/metabolism , Homeostasis/physiology , Receptors, Purinergic P2/physiology , Renal Circulation/physiology , Adenosine Triphosphate/pharmacology , Analysis of Variance , Animals , Dogs , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Homeostasis/drug effects , Injections, Intra-Arterial , Kidney Function Tests , Nitroarginine/pharmacology , Receptors, Purinergic P2/drug effects , Reference Values , Renal Circulation/drug effects , Vascular Resistance/drug effects , Vascular Resistance/physiology
18.
Am J Physiol ; 276(3): F450-6, 1999 03.
Article in English | MEDLINE | ID: mdl-10070169

ABSTRACT

We performed studies to determine the effect of extracellular ATP on the intracellular Ca2+ concentration ([Ca2+]i) in freshly isolated microvascular smooth muscle cells (MVSMC). Suspensions of preglomerular MVSMC were prepared by enzymatic digestion and loaded with fura 2. Single cells were studied using a microscope-based fluorescence spectrophotometer during superfusion of a physiological salt solution with 1.8 mM Ca2+ and during exposure to similar solutions containing ATP. Under control conditions, baseline [Ca2+]i averaged 107 +/- 6 nM (n = 86 cells from 34 animals). ATP administration elicited concentration-dependent increases in [Ca2+]i. Exposure to ATP concentrations of 1, 10, and 100 microM increased intracellular Ca2+ to peak concentrations of 133 +/- 20, 338 +/- 37, and 367 +/- 35 nM, respectively (P < 0.05 vs. respective baseline). Steady-state [Ca2+]i increased to 113 +/- 15, 150 +/- 16 (P < 0.05 vs. baseline), and 180 +/- 12 nM (P < 0.05 vs. baseline) for the same groups. The [Ca2+]i response to ATP was also assessed in the absence of extracellular Ca2+ and during blockade of L-type Ca2+ channels with diltiazem. In these studies, exposure to 100 microM ATP induced a transient peak increase in [Ca2+]i with the plateau phase being totally abolished under Ca2+-free conditions and markedly attenuated during Ca2+ channel blockade, respectively. These data indicate that ATP-mediated P2-receptor activation increases [Ca2+]i in freshly isolated preglomerular MVSMC by stimulating Ca2+ release from intracellular stores, in addition to stimulating the influx of extracellular Ca2+ through voltage-gated L-type Ca2+ channels.


Subject(s)
Adenosine Triphosphate/pharmacology , Calcium Signaling/drug effects , Kidney Glomerulus/blood supply , Muscle, Smooth, Vascular/drug effects , Animals , Arteries/cytology , Arteries/drug effects , Arteries/physiology , Arterioles/cytology , Arterioles/drug effects , Arterioles/physiology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels, L-Type , Calcium Signaling/physiology , Diltiazem/pharmacology , Dose-Response Relationship, Drug , Homeostasis/drug effects , In Vitro Techniques , Intracellular Membranes/metabolism , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Osmolar Concentration , Rats , Rats, Sprague-Dawley
19.
Hypertension ; 33(1 Pt 2): 195-200, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9931104

ABSTRACT

-The current studies were performed to determine the contribution of calcium mobilization and voltage-dependent calcium influx to the increase in [Ca2+]i elicited by ATP and UTP. Suspensions of freshly isolated smooth muscle cells were prepared from preglomerular microvessels by enzymatic digestion and loaded with the Ca2+-sensitive dye fura 2. The effect of ATP and UTP on [Ca2+]i was studied on single cells with standard microscope-based fluorescence photometry techniques. Resting [Ca2+]i averaged 80+/-3 nmol/L (n=219 single cells from 58 dispersions). ATP (100 micromol/L) increased [Ca2+]i to a peak value of 845+/-55 nmol/L (n=70 single cells from 38 dispersions) before stabilizing at 124+/-81 nmol/L. Similarly, 100 micromol/L UTP (n=39 single cells from 26 dispersions) stimulated a peak increase in [Ca2+]i of 1426+/-584 nmol/L before reaching a stable plateau of 123+/-10 nmol/L. The [Ca2+]i response to ATP and UTP was also assessed in the absence of extracellular calcium. In these studies, exposure to 100 micromol/L ATP induced a transient peak increase in [Ca2+]i, with the plateau phase being totally abolished. In contrast, exposure to 100 micromol/L UTP under calcium-free conditions resulted in no detectable change in the UTP-mediated increase in [Ca2+]i. The role of L-type calcium channels in the response was assessed with the calcium channel antagonist diltiazem. Incubation with diltiazem (10 micromol/L) markedly reduced the response to ATP, whereas the response to UTP was only slightly reduced. These data demonstrate that both ATP and UTP directly stimulate a biphasic increase in [Ca2+]i in renal microvascular smooth muscle cells. Furthermore, the data suggest that the elevation of [Ca2+]i elicited by ATP is largely dependent on calcium influx through L-type calcium channels, whereas the response to UTP appears to derive primarily from mobilization of calcium from intracellular stores.


Subject(s)
Adenosine Triphosphate/physiology , Calcium/metabolism , Kidney Cortex/blood supply , Microcirculation/physiology , Muscle, Smooth, Vascular/physiology , Receptors, Purinergic/physiology , Signal Transduction , Adenosine Triphosphate/pharmacology , Animals , Cells, Cultured , Kinetics , Male , Microcirculation/drug effects , Muscle, Smooth, Vascular/drug effects , Rats , Rats, Sprague-Dawley , Uridine Triphosphate/pharmacology , Uridine Triphosphate/physiology
20.
Hypertension ; 33(1 Pt 2): 408-13, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9931138

ABSTRACT

The current study determined the contribution of protein kinase-A (PKA) and protein kinase-G (PKG) to the vasodilation elicited by the N-methylsulfonimide analog of 11,12-epoxyeicosatrienoic acid (11, 12-EET). Experiments were performed, in vitro, using the juxtamedullary nephron preparation combined with videomicroscopy. The response of afferent arterioles to the sulfonimide analog of 11, 12-EET, was determined before and after inhibition of PKA, PKG, or guanylyl cyclase. Afferent arterioles, preconstricted with 0.5 micromol/L norepinephrine, averaged 18+/-1 microm (n=25) at a renal perfusion pressure of 100 mm Hg. Superfusion with 0.01 to 100 nmol/L of the 11,12-EET analog caused a graded increase in diameter of the afferent arteriole. Vessel diameter increased by 11+/-1% and 15+/-1%, respectively, in response to 10 and 100 nmol/L of the 11,12-EET analog. The afferent arteriolar response to 10 and 100 nmol/L of the 11,12-EET analog was significantly attenuated during inhibition of PKA with 10 micromol/L H-89 (n=7) or 5 micromol/L myristolated PKI (n=6), such that afferent arteriolar diameter increased by only 5+/-2% and 2+/-1%, respectively, in response to 100 nmol/L of the 11, 12-EET analog. In contrast, the afferent arteriolar vasodilatory response to the 11,12-EET analog was unaffected by PKG or guanylyl cyclase inhibition. In the presence of 200 micromol/L histone H2B (n=5) or 10 micromol/L ODQ (n=7), the afferent arteriolar diameter increased by 16+/-3% and 12+/-2%, respectively, in response to 100 nmol/L of the 11,12-EET analog. These results demonstrate that activation of PKA is an important mechanism responsible for the afferent arteriolar vasodilation elicited by the sulfonimide analog of 11,12-EET.


Subject(s)
8,11,14-Eicosatrienoic Acid/pharmacology , Arterioles/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Muscle, Smooth, Vascular/physiology , Renal Circulation/drug effects , Sulfonamides , Vasodilation/drug effects , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Arterioles/drug effects , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases , Guanylate Cyclase/metabolism , Histones/pharmacology , In Vitro Techniques , Isoquinolines/pharmacology , Juxtaglomerular Apparatus/blood supply , Kidney Medulla/blood supply , Kinetics , Male , Microscopy, Video , Muscle, Smooth, Vascular/drug effects , Nephrons/blood supply , Norepinephrine/pharmacology , Oxadiazoles/pharmacology , Protein Kinases/metabolism , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...