Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 264
Filter
1.
Am J Physiol Renal Physiol ; 315(3): F521-F534, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29667908

ABSTRACT

The prorenin receptor (PRR) was originally proposed to be a member of the renin-angiotensin system (RAS); however, recent work questioned their association. The present paper describes a functional link between the PRR and RAS in the renal juxtaglomerular apparatus (JGA), a classic anatomical site of the RAS. PRR expression was found in the sensory cells of the JGA, the macula densa (MD), and immunohistochemistry-localized PRR to the MD basolateral cell membrane in mouse, rat, and human kidneys. MD cell PRR activation led to MAP kinase ERK1/2 signaling and stimulation of PGE2 release, the classic pathway of MD-mediated renin release. Exogenous renin or prorenin added to the in vitro microperfused JGA-induced acute renin release, which was inhibited by removing the MD or by the administration of a PRR decoy peptide. To test the function of MD PRR in vivo, we established a new mouse model with inducible conditional knockout (cKO) of the PRR in MD cells based on neural nitric oxide synthase-driven Cre-lox recombination. Deletion of the MD PRR significantly reduced blood pressure and plasma renin. Challenging the RAS by low-salt diet + captopril treatment caused further significant reductions in blood pressure, renal renin, cyclooxygenase-2, and microsomal PGE synthase expression in cKO vs. wild-type mice. These results suggest that the MD PRR is essential in a novel JGA short-loop feedback mechanism, which is integrated within the classic MD mechanism to control renin synthesis and release and to maintain blood pressure.


Subject(s)
Blood Pressure , Juxtaglomerular Apparatus/enzymology , Proton-Translocating ATPases/metabolism , Receptors, Cell Surface/metabolism , Renin-Angiotensin System , Renin/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Biosensing Techniques , Blood Pressure/drug effects , Captopril/pharmacology , Cyclooxygenase 2/metabolism , Diet, Sodium-Restricted , Dinoprostone/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Juxtaglomerular Apparatus/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Prostaglandin-E Synthases/metabolism , Proton-Translocating ATPases/deficiency , Proton-Translocating ATPases/genetics , Rats, Sprague-Dawley , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Renin-Angiotensin System/drug effects , Secretory Pathway , Signal Transduction , Vacuolar Proton-Translocating ATPases/genetics , Prorenin Receptor
2.
High Blood Press Cardiovasc Prev ; 24(3): 231-242, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28527017

ABSTRACT

Renin is the first and rate-limiting step of the renin-angiotensin system. The exclusive source of renin in the circulation are the juxtaglomerular cells of the kidney, which line the afferent arterioles at the entrance of the glomeruli. Normally, renin production by these cells suffices to maintain homeostasis. However, under chronic stimulation of renin release, for instance during a low-salt diet or antihypertensive therapy, cells that previously expressed renin during congenital life re-convert to a renin-producing cell phenotype, a phenomenon which is known as "recruitment". How exactly such differentiation occurs remains to be clarified. This review critically discusses the phenotypic plasticity of renin cells, connecting them not only to the classical concept of blood pressure regulation, but also to more complex contexts such as development and growth processes, cell repair mechanisms and tissue regeneration.


Subject(s)
Cardiovascular Diseases/metabolism , Cell Plasticity , Juxtaglomerular Apparatus/metabolism , Kidney Diseases/metabolism , Renin-Angiotensin System , Animals , Calcium Signaling , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cell Plasticity/drug effects , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Humans , Juxtaglomerular Apparatus/drug effects , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Phenotype , Regeneration , Renin-Angiotensin System/drug effects , Second Messenger Systems
3.
Hypertension ; 65(5): 1047-54, 2015 May.
Article in English | MEDLINE | ID: mdl-25776075

ABSTRACT

Despite the importance of juxtaglomerular cell recruitment in the pathophysiology of cardiovascular diseases, the mechanisms that underlie renin production under conditions of chronic stimulation remain elusive. We have previously shown that CD44+ mesenchymal-like cells (CD44+ cells) exist in the adult kidney. Under chronic sodium deprivation, these cells are recruited to the juxtaglomerular area and differentiate to new renin-expressing cells. Given the proximity of macula densa to the juxtaglomerular area and the importance of macula densa released prostanoids in renin synthesis and release, we hypothesized that chronic sodium deprivation induces macula densa release of prostanoids, stimulating renal CD44+ cell activation and differentiation. CD44+ cells were isolated from adult kidneys and cocultured with the macula densa cell line, MMDD1, in normal or low-sodium medium. Low sodium stimulated prostaglandin E2 production by MMDD1 and induced migration of CD44+ cells. These effects were inhibited by addition of a cyclooxygenase 2 inhibitor (NS398) or an E-prostanoid receptor 4 antagonist (AH23848) to MMDD1 or CD44+ cells, respectively. Addition of prostaglandin E2 to CD44+ cells increased cell migration and induced renin expression. In vivo activation of renal CD44+ cells during juxtaglomerular recruitment was attenuated in wild-type mice subjected to salt restriction in the presence of cyclooxygenase 2 inhibitor rofecoxib. Similar results were observed in E-prostanoid receptor 4 knockout mice subjected to salt restriction. These results show that the prostaglandin E2/E-prostanoid receptor 4 pathway plays a key role in the activation of renal CD44+ mesenchymal stromal cell-like cells during conditions of juxtaglomerular recruitment; highlighting the importance of this pathway as a key regulatory mechanism of juxtaglomerular recruitment.


Subject(s)
Dinoprostone/genetics , Gene Expression Regulation , Hypertension/diet therapy , Mesenchymal Stem Cells/physiology , RNA, Messenger/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Animals , Cell Differentiation , Cell Line , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/biosynthesis , Disease Models, Animal , Hypertension/genetics , Hypertension/metabolism , Immunoblotting , Immunohistochemistry , Juxtaglomerular Apparatus/drug effects , Juxtaglomerular Apparatus/metabolism , Juxtaglomerular Apparatus/pathology , Male , Mice , Mice, Inbred C57BL , Receptors, Prostaglandin E, EP4 Subtype/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
4.
Am J Physiol Renal Physiol ; 306(8): F864-72, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24431205

ABSTRACT

Iodinated contrast media (CM) have adverse effects that may result in contrast-induced acute kidney injury. Oxidative stress is believed to play a role in CM-induced kidney injury. We test the hypothesis that oxidative stress and reduced nitric oxide in tubules are consequences of CM-induced direct cell damage and that increased local oxidative stress may increase tubuloglomerular feedback. Rat thick ascending limbs (TAL) were isolated and perfused. Superoxide and nitric oxide were quantified using fluorescence techniques. Cell death rate was estimated using propidium iodide and trypan blue. The function of macula densa and tubuloglomerular feedback responsiveness were measured in isolated, perfused juxtaglomerular apparatuses (JGA) of rabbits. The expression of genes related to oxidative stress and the activity of superoxide dismutase (SOD) were investigated in the renal medulla of rats that received CM. CM increased superoxide concentration and reduced nitric oxide bioavailability in TAL. Propidium iodide fluorescence and trypan blue uptake increased more in CM-perfused TAL than in controls, indicating increased rate of cell death. There were no marked acute changes in the expression of genes related to oxidative stress in medullary segments of Henle's loop. SOD activity did not differ between CM and control groups. The tubuloglomerular feedback in isolated JGA was increased by CM. Tubular cell damage and accompanying oxidative stress in our model are consequences of CM-induced direct cell damage, which also modifies the tubulovascular interaction at the macula densa, and may therefore contribute to disturbances of renal perfusion and filtration.


Subject(s)
Contrast Media/adverse effects , Juxtaglomerular Apparatus/drug effects , Kidney Tubules/drug effects , Loop of Henle/drug effects , Triiodobenzoic Acids/adverse effects , Acute Kidney Injury/chemically induced , Acute Kidney Injury/physiopathology , Animals , Biological Availability , Cell Death/drug effects , Feedback, Physiological/drug effects , In Vitro Techniques , Juxtaglomerular Apparatus/physiology , Kidney Tubules/metabolism , Loop of Henle/metabolism , Male , Nitric Oxide/metabolism , Nitric Oxide/pharmacokinetics , Oxidative Stress/drug effects , Perfusion , Rabbits , Rats , Superoxides/metabolism , Transcriptome/drug effects
5.
Article in English | MEDLINE | ID: mdl-23500064

ABSTRACT

Previous studies have indicated that 20-hydroxyeicosatetraeonic acid (20-HETE) modulates vascular tone in large cerebral and renal arteries through inhibition of the large conductance, calcium sensitive potassium (BK) channel activity. However, the role of 20-HETE in modulating tubuloglomerular feedback (TGF) and the myogenic response in the afferent arteriole (Af-Art) is unknown. The present study examined the effects of inhibitors of the synthesis and action of 20-HETE on the myogenic and TGF responses of isolated rabbit and mouse Af-Arts. Luminal diameter decreased by 9.2±0.5% in mice and 8.9±1.3% in rabbit Af-Art when the perfusion pressure was increased from 60 to 120 mmHg. Administration of a 20-HETE synthesis inhibitor, HET0016 (1 µM), or a selective 20-HETE antagonist, 6, 15-20-hydroxyeicosadienoic acid (6, 15-20-HEDE, 10 µM) completely blocked the myogenic response of both rabbit and mouse Af-Art, while addition of 5, 14-20-HEDE (10 µM), a 20-HETE agonist, restored the myogenic response in vessels treated with HET0016. Increases in NaCl concentration from 10 to 80 mM of the solution perfusing the macula densa constricted the Af-Art of rabbits by 6.0±1.4 µm (n=5). Addition of a 20-HETE agonist to the tubular perfusate potentiated the TGF-mediated vasoconstrictor response. This response was blocked by addition of a 20-HETE antagonist (6, 15-20-HEDE, 10 µM) to the vascular perfusate. These studies indicate that locally produced 20-HETE plays an important role in modulating the myogenic and TGF responsiveness of the Af-Art and may help explain how deficiencies in the renal formation of 20-HETE could promote the development of hypertension induced glomerular injury.


Subject(s)
Arterioles/drug effects , Hydroxyeicosatetraenoic Acids/administration & dosage , Hypertension/physiopathology , Juxtaglomerular Apparatus/drug effects , Kidney Tubules/blood supply , Animals , Arachidonic Acid/administration & dosage , Arachidonic Acid/metabolism , Arterioles/physiology , Blood Pressure/drug effects , Humans , Hypertension/drug therapy , Juxtaglomerular Apparatus/blood supply , Kidney Glomerulus/blood supply , Kidney Glomerulus/drug effects , Kidney Tubules/drug effects , Mice , Microvessels/metabolism , Microvessels/pathology , Perfusion , Rabbits
6.
Am J Physiol Renal Physiol ; 304(5): F498-504, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23269646

ABSTRACT

Renin, the rate-limiting enzyme in the formation of angiotensin II, is synthesized and stored in granules in juxtaglomerular (JG) cells. Therefore, the controlled mechanism involved in renin release is essential for the regulation of blood pressure. Exocytosis of renin-containing granules is likely involved in renin release; a process stimulated by cAMP. We found that the "soluble NSF (N-ethylmaleimide-sensitive factor) attachment protein receptor" (SNARE) protein VAMP2 mediates cAMP-stimulated renin release and exocytosis in JG cells. To mediate exocytosis, VAMP2 must interact with a synaptosome-associated protein (SNAP). In the renal cortex, the isoform SNAP23 is abundantly expressed. We hypothesized that SNAP23 mediates cAMP-stimulated renin release from primary cultures of mouse JG cells. We found that SNAP23 protein is expressed and colocalized with renin-containing granules in primary cultures of mouse JG cell lysates. Thus, we then tested the involvement of SNAP23 in cAMP-stimulated renin release by transducing JG cells with a dominant-negative SNAP23 construct. In control JG cells transduced with a scrambled sequence, increasing cAMP stimulated renin release from 1.3 ± 0.3 to 5.3 ± 1.2% of renin content. In cells transduced with dominant-negative SNAP23, cAMP increased renin from 1.0 ± 0.1 to 3.0 ± 0.6% of renin content, a 50% blockade. Botulinum toxin E, which cleaves and inactivates SNAP23, reduced cAMP-stimulated renin release by 42 ± 17%. Finally, adenovirus-mediated silencing of SNAP23 significantly blocked cAMP-stimulated renin release by 50 ± 13%. We concluded that the SNARE protein SNAP23 mediates cAMP-stimulated renin release. These data show that renin release is a SNARE-dependent process.


Subject(s)
Cyclic AMP/metabolism , Juxtaglomerular Apparatus/metabolism , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Renin/metabolism , Animals , Cells, Cultured , Colforsin/pharmacology , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/drug effects , Mice , Qb-SNARE Proteins/genetics , Qc-SNARE Proteins/genetics
7.
Am J Physiol Renal Physiol ; 304(3): F248-56, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23220722

ABSTRACT

The calcium-sensing receptor (CaSR) is a G-coupled protein expressed in renal juxtaglomerular (JG) cells. Its activation stimulates calcium-mediated decreases in cAMP content and inhibits renin release. The postreceptor pathway for the CaSR in JG cells is unknown. In parathyroids, CaSR acts through G(q) and/or G(i). Activation of G(q) stimulates phospholipase C (PLC), and inositol 1,4,5-trisphosphate (IP(3)), releasing calcium from intracellular stores. G(i) stimulation inhibits cAMP formation. In afferent arterioles, the ryanodine receptor (RyR) enhances release of stored calcium. We hypothesized JG cell CaSR activation inhibits renin via the PLC/IP(3) and also RyR activation, increasing intracellular calcium, suppressing cAMP formation, and inhibiting renin release. Renin release from primary cultures of isolated mouse JG cells (n = 10) was measured. The CaSR agonist cinacalcet decreased renin release 56 ± 7% of control (P < 0.001), while the PLC inhibitor U73122 reversed cinacalcet inhibition of renin (104 ± 11% of control). The IP(3) inhibitor 2-APB also reversed inhibition of renin from 56 ± 6 to 104 ± 11% of control (P < 0.001). JG cells were positively labeled for RyR, and blocking RyR reversed CaSR-mediated inhibition of renin from 61 ± 8 to 118 ± 22% of control (P < 0.01). Combining inhibition of IP(3) and RyR was not additive. G(i) inhibition with pertussis toxin plus cinacalcet did not reverse renin inhibition (65 ± 12 to 41 ± 8% of control, P < 0.001). We conclude stimulating JG cell CaSR activates G(q), initiating the PLC/IP(3) pathway, activating RyR, increasing intracellular calcium, and resulting in calcium-mediated renin inhibition.


Subject(s)
Inositol 1,4,5-Trisphosphate/metabolism , Juxtaglomerular Apparatus/metabolism , Receptors, Calcium-Sensing/metabolism , Renin/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction/physiology , Type C Phospholipases/metabolism , Animals , Calcimimetic Agents/pharmacology , Calcium/metabolism , Cells, Cultured , Cinacalcet , Cyclic AMP/metabolism , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/drug effects , Mice , Models, Animal , Naphthalenes/pharmacology , Pertussis Toxin/pharmacology , Pyrrolidinones/pharmacology , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/drug effects , Type C Phospholipases/antagonists & inhibitors
8.
Am J Physiol Renal Physiol ; 303(8): F1157-65, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22896038

ABSTRACT

Parathyroid hormone (PTH) is positively coupled to the generation of cAMP via its actions on the PTH1R and PTH2R receptors. Renin secretion from juxtaglomerular (JG) cells is stimulated by elevated intracellular cAMP, and every stimulus that increases renin secretion is thought to do so via increasing cAMP. Thus we hypothesized that PTH increases renin release from primary cultures of mouse JG cells by elevating intracellular cAMP via the PTH1R receptor. We found PTH1R, but not PTH2R, mRNA expressed in JG cells. While PTH increased JG cell cAMP content from (log(10) means ± SE) 3.27 ± 0.06 to 3.92 ± 0.12 fmol/mg protein (P < 0.001), it did not affect renin release. The PTH1R-specific agonist, parathyroid hormone-related protein (PTHrP), also increased JG cell cAMP from 3.13 ± 0.09 to 3.93 ± 0.09 fmol/mg protein (P < 0.001), again without effect on renin release. PTH2R receptor agonists had no effect on cAMP or renin release. PTHrP increased cAMP in the presence of both low and high extracellular calcium from 3.31 ± 0.17 to 3.83 ± 0.20 fmol/mg protein (P < 0.01) and from 3.29 ± 0.18 to 3.63 ± 0.22 fmol/mg protein (P < 0.05), respectively, with no effect on renin release. PTHrP increased JG cell cAMP in the presence of adenylyl cyclase-V inhibition from 2.85 ± 0.17 to 3.44 ± 0.14 fmol/mg protein (P < 0.001) without affecting renin release. As a positive control, forskolin increased JG cell cAMP from 3.39 ± 0.13 to 4.48 ± 0.07 fmol/mg protein (P < 0.01) and renin release from 2.96 ± 0.10 to 3.29 ± 0.08 ng ANG I·mg prot(-1)·h(-1) (P < 0.01). Thus PTH increases JG cell cAMP via non-calcium-sensitive adenylate cyclases without affecting renin release. These data suggest compartmentalization of cAMP signaling in JG cells.


Subject(s)
Cyclic AMP/metabolism , Juxtaglomerular Apparatus/drug effects , Parathyroid Hormone/pharmacology , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptor, Parathyroid Hormone, Type 2/metabolism , Renin/metabolism , Animals , Cells, Cultured , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/metabolism , Mice , Parathyroid Hormone/metabolism
9.
BMC Nephrol ; 13: 21, 2012 Apr 25.
Article in English | MEDLINE | ID: mdl-22533828

ABSTRACT

BACKGROUND: Dual renin-angiotensin system blockade with angiotensin-converting enzyme inhibitors and angiotensin receptor blockers has been advocated to minimize proteinuria. However, recent trials have questioned the renal safety of this approach. Our understanding on the molecular effects of dual blockade in humans is incomplete. CASE PRESENTATION: We present a patient with corticoid resistant nephrotic syndrome who developed marked juxtaglomerular apparatus hyperplasia and renin expression in the context of dual angiotensin system blockade. CONCLUSIONS: Although renin may have profibrotic effects mediated by (pro)renin receptor activation, this report raises questions on the potential consequences of local renin activation on chronic kidney disease in patients with dual angiotensin blockade.


Subject(s)
Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Juxtaglomerular Apparatus/pathology , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/pathology , Renin-Angiotensin System , Angiotensin Receptor Antagonists/adverse effects , Angiotensin Receptor Antagonists/pharmacology , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Humans , Hyperplasia , Juxtaglomerular Apparatus/drug effects , Male , Nephrotic Syndrome/metabolism , Renin/antagonists & inhibitors , Renin/biosynthesis , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Young Adult
10.
Hypertension ; 59(3): 599-606, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22311906

ABSTRACT

Chronic aldosterone administration increases glomerular filtration rate, whereas inhibition of mineralocorticoid receptors (MRs) markedly attenuates glomerular hyperfiltration and hypertension associated with primary aldosteronism or obesity. However, the mechanisms by which aldosterone alters glomerular filtration rate regulation are poorly understood. In the present study, we hypothesized that aldosterone suppresses tubuloglomerular feedback (TGF) via activation of macula densa MR. First, we observed the expression of MR in macula densa cells isolated by laser capture microdissection and by immunofluorescence in rat kidneys. Second, to investigate the effects of aldosterone on TGF in vitro, we microdissected the juxtaglomerular apparatus from rabbit kidneys and perfused the afferent arteriole and distal tubule simultaneously. Under control conditions, TGF was 2.8±0.2 µm. In the presence of aldosterone (10(-8) mol/L), TGF was reduced by 50%. The effect of aldosterone to attenuate TGF was blocked by the MR antagonist eplerenone (10(-5) mol/L). Third, to investigate the effect of aldosterone on TGF in vivo, we performed micropuncture, and TGF was determined by maximal changes in stop-flow pressure P(sf) when tubular perfusion rate was increased from 0 to 40 nL/min. Aldosterone (10(-7) mol/L) decreased ΔP(sf) from 10.1±1.4 to 7.7±1.2 mm Hg. In the presence of l-NG-monomethyl arginine citrate (10(-3) mol/L), this effect was blocked. We conclude that MRs are expressed in macula densa cells and can be activated by aldosterone, which increases nitric oxide production in the macula densa and blunts the TGF response.


Subject(s)
Aldosterone/pharmacology , Glomerular Filtration Rate/drug effects , Juxtaglomerular Apparatus/metabolism , Kidney Tubules, Distal/metabolism , Receptors, Mineralocorticoid/metabolism , Animals , Cells, Cultured , Feedback/drug effects , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/drug effects , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/drug effects , Male , Nitric Oxide/biosynthesis , Rabbits , Rats , Rats, Sprague-Dawley , Receptors, Mineralocorticoid/drug effects
11.
Oncol Rep ; 27(3): 842-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22024718

ABSTRACT

A stable nitroxide 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-osyl (Tempol) is widely used as an antioxidant in vitro and in vivo. In this study, we investigated the effects of Tempol on the growth of As4.1 juxtaglomerular cells in relation to cell cycle and cell death. Tempol dose-dependently decreased the growth of As4.1 cells with an IC50 of ~1 mM at 48 h. DNA flow cytometry analysis and BrdU staining indicated that Tempol induced S phase arrest, which is accompanied by a downregulation of cyclin A. Tempol also induced apoptotic cell death, which was accompanied by the loss of mitochondrial membrane potential (MMP; ∆Ψm), an activation of caspase-3 and cleavage of poly(ADP-ribose)polymerase-1 (PARP-1). Furthermore, Tempol increased reactive oxygen species (ROS) levels, and the phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK). MEK and JNK inhibitors significantly attenuated a growth inhibition in Tempol-treated As4.1 cells. In conclusion, Tempol inhibited the growth of As4.1 cells via cell cycle arrest and apoptosis. Tempol also activated ERK and JNK signaling, which was responsible for cell growth inhibition. Our present data provide useful information for the toxicological effects of Tempol in juxtaglomerular cells in relation to cell growth inhibition and cell death.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cyclic N-Oxides/pharmacology , Juxtaglomerular Apparatus/drug effects , Animals , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cyclin A/metabolism , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutathione/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Phosphorylation/drug effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , S Phase/drug effects , Signal Transduction/drug effects , Spin Labels
12.
Nephron Exp Nephrol ; 122(3-4): 83-94, 2012.
Article in English | MEDLINE | ID: mdl-23548923

ABSTRACT

BACKGROUND/AIMS: Pioglitazone (PGZ), one of the thiazolidinediones, has been known to show renoprotective effects. In this study, we focused on the effect of PGZ on glomerular hyperfiltration (GHF), resultant glomerular injury and altered macula densa signaling as a cause of sustained GHF through modified tubuloglomerular feedback in rats with diabetic nephropathy. METHODS: Kidneys from 24-week-old male OLETF rats and LET rats, nondiabetic controls, were used for the experiment. PGZ was administered (10 mg/kg/day, p.o.) for 2 weeks from 22 to 24 weeks of age in some of the OLETF rats (OLETF+PGZ). RESULTS: Parameters relating GHF, kidney weight, creatinine clearance, urine albumin/creatinine ratio and glomerular surface were all increased in OLETF rats and partially restored in OLETF+PGZ rats. Expressions of desmin and TGF-ß were also increased in OLETF rats and restored in OLETF+PGZ rats. The changes in TGF-ß expression were confirmed to be independent of podocyte number. Finally, the immunoreactivity of neuronal nitric oxide synthase (nNOS) and cyclooxygenase 2 (COX-2) in the macula densa was assessed for the evaluation of macula densa signaling. Altered intensities of nNOS and COX-2 in OLETF rats were restored in OLETF+PGZ rats, which agreed with the gene expression analysis (nNOS: 100.2 ± 2.9% in LET, 64.2 ± 2.7% in OLETF, 87.4 ± 12.1% in OLETF+PGZ; COX-2: 100.8 ± 7.4% in LET, 249.2 ± 19.4% in OLETF, 179.9 ± 13.5% in OLETF+PGZ; n = 5) and the semiquantitative analysis of nNOS/COX-2-positive cells. CONCLUSION: PGZ effectively attenuated the GHF and hyperfiltration-associated glomerular injury in diabetic nephropathy. The restoration of altered macula densa signaling might be involved in the renoprotective effect of PGZ.


Subject(s)
Diabetic Nephropathies/drug therapy , Juxtaglomerular Apparatus/physiology , Kidney Glomerulus/drug effects , Thiazolidinediones/therapeutic use , Animals , Cyclooxygenase 2/metabolism , Desmin/biosynthesis , Diabetic Nephropathies/prevention & control , Juxtaglomerular Apparatus/drug effects , Male , Nitric Oxide Synthase Type I/metabolism , Pioglitazone , Rats , Rats, Inbred OLETF , Signal Transduction , Thiazolidinediones/pharmacology , Transforming Growth Factor beta/biosynthesis
13.
Hypertension ; 58(4): 611-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21859963

ABSTRACT

Proteinase-activated receptors (PARs) 1 to 4 are highly expressed in the kidney and are involved in the regulation of renal hemodynamics and tubular function. Since intravascular infusion of the proteinase thrombin, which activates PARs, has been shown to decrease plasma renin activity in rats, we investigated the effects of the respective PAR subtypes on renin release using the isolated perfused mouse kidney model. Thrombin dose-dependently reduced perfusate flow and inhibited renin secretion rates (RSRs) that had been prestimulated by the ß-adrenoreceptor agonist isoproterenol. The suppression of RSRs was prevented by the selective PAR1 inhibitor SCH79797, and direct activation of PAR1 by TFLLR mimicked the effects of thrombin on RSRs and vascular tone. Moreover, TFLLR suppressed the stimulations of RSRs in response to the loop diuretic bumetanide, to prostaglandin E(2), or to a decrease in renal perfusion pressure but not in response to a reduction in extracellular calcium. The PAR2-activating peptide SLIGRL concentration dependently increased RSR and perfusate flow. The stimulation of RSRs by SLIGRL was markedly attenuated by N(G)-nitro-L-arginine methyl ester, suggesting an NO-dependent mechanism. Activation of PAR4 by AYPGKF did not modulate RSRs or perfusate flow. PAR1 and PAR2 immunoreactivity were detected in the juxtaglomerular region and were colocalized with renin immunoreactivity. Our data provide evidence that PAR1 activation inhibits renal renin secretion and induces renal vasoconstriction, whereas PAR2 activation stimulates renin release and induces vasodilation mainly via the release of NO.


Subject(s)
Juxtaglomerular Apparatus/metabolism , Kidney/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Renin/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Dose-Response Relationship, Drug , Isoproterenol/pharmacology , Juxtaglomerular Apparatus/drug effects , Kidney/drug effects , Male , Mice , Mice, Inbred C57BL , Models, Animal , Nitric Oxide/metabolism , Oligopeptides/pharmacology , Pyrroles/pharmacology , Quinazolines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/drug effects , Receptor, PAR-2/drug effects , Thrombin/pharmacology
14.
Hypertens Res ; 34(8): 942-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21633358

ABSTRACT

Calcium (Ca(2+)) has an important role in nearly all types of cellular secretion, with a particularly novel role in the juxtaglomerular (JG) cells in the kidney. In JG cells, Ca(2+) inhibits renin secretion, which is a major regulator of blood pressure and renal hemodynamics. However, whether alterations in afferent arteriolar (Af-Art) pressure change intracellular Ca(2+) concentration ([Ca(2+)](i)) in JG cells and whether [Ca(2+)](i) comes from extracellular or intracellular sources remains unknown. We hypothesize that increases in perfusion pressure in the Af-Art result in elevations in [Ca(2+)](i) in JG cells. We isolated and perfused Af-Art of C57BL6 mice and measured changes in [Ca(2+)](i) in JG cells in response to perfusion pressure changes. The JG cells' [Ca(2+)](i) was 93.3±2.2 nM at 60 mm Hg perfusion pressure and increased to 111.3±13.4, 119.6±7.3, 130.3±2.9 and 140.8±12.1 nM at 80, 100, 120 and 140 mm Hg, respectively. At 120 mm Hg, increases in [Ca(2+)](i) were reduced in mice receiving the following treatments: (1) the mechanosensitive cation channel blocker, gadolinium (94.6±7.5 nM); (2) L-type calcium channel blocker, nifedipine (105.8±7.5 nM); and (3) calcium-free solution plus ethylene glycol tetraacetic acid (96.0±5.8 nM). Meanwhile, the phospholipase C inhibitor, inositol triphosphate receptor inhibitor, T-type calcium channel blocker, N-type calcium channel blocker and Ca(2+)-ATPase inhibitor did not influence changes in [Ca(2+)](i) in JG cells. In summary, JG cell [Ca(2+)](i) rise as perfusion pressure increases; furthermore, the calcium comes from extracellular sources, specifically mechanosensitive cation channels and L-type calcium channels.


Subject(s)
Arterioles/metabolism , Calcium/metabolism , Juxtaglomerular Apparatus/metabolism , Kidney/blood supply , Animals , Arterioles/cytology , Arterioles/drug effects , Calcium Channel Blockers/pharmacology , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Gadolinium/pharmacology , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/drug effects , Kidney/drug effects , Kidney/metabolism , Male , Mice , Nifedipine/pharmacology , Pressure
15.
Int J Mol Med ; 27(4): 575-81, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21258765

ABSTRACT

Carbonyl cyanide p-(trifluoromethoxy) phenylhydrazone (FCCP) is an uncoupler of mitochondrial oxidative phosphorylation in eukaryotic cells. In the present study, we investigated the involvement of reactive oxygen species (ROS) and glutathione (GSH) in FCCP-induced As4.1 juxtaglomerular cell death. Intracellular ROS levels were decreased by FCCP at the early time points (10-150 min) and increased at 48 h. FCCP inhibited the activity of Mn-superoxide dismutase (Mn-SOD) via down-regulating its protein expression. Ebselen (an antioxidant) significantly attenuated ROS levels in FCCP-treated cells, but did not prevent FCCP-induced cell death. Moreover, intracellular GSH content was rapidly diminished within 10 min of FCCP treatment, which was accompanied by a reduction of the mitochondrial membrane potential [MMP (∆ψm)]. L-buthionine sulfoximine (BSO, a GSH synthesis inhibitor) significantly augmented As4.1 cell death by FCCP. However, N-acetylcysteine (NAC, a GSH precursor and antioxidant) attenuated GSH depletion, MMP (∆ψm) loss and cell death in FCCP-treated As4.1 cells. In addition, NAC increased Mn-SOD activity and decreased ROS levels in FCCP-treated As4.1 cells. In conclusion, these results suggest that compared to ROS levels, intracellular GSH levels are more closely linked to FCCP-induced apoptosis in As4.1 juxtaglomerular cells.


Subject(s)
Apoptosis/drug effects , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Glutathione/metabolism , Intracellular Space/metabolism , Juxtaglomerular Apparatus/drug effects , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Animals , Buthionine Sulfoximine/metabolism , Buthionine Sulfoximine/pharmacology , Catalase/metabolism , Cell Death/drug effects , Cell Line , Juxtaglomerular Apparatus/metabolism , Mice , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
16.
Anticancer Res ; 30(7): 2863-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20683024

ABSTRACT

Carbonyl cyanide p-(trifluoromethoxy) phenylhydrazone (FCCP) is an uncoupler of mitochondrial oxidative phosphorylation in mitochondria. This study evaluated the effects of FCCP on the growth of juxtaglomerular As4.1 cells in relation to the cell cycle and apoptosis. FCCP inhibited the growth of As4.1 cells with an IC(50) of approximately 10 muM at 48 hours. DNA flow cytometry indicated that FCCP did not induce the specific phase arrests of the cell cycle. This agent efficiently reduced mitochondrial membrane potential (MMP; DeltaPsi(m)) levels within 1 hour and dose-dependently induced loss of MMP (DeltaPsi(m)) at 48 h. FCCP also induced apoptosis in As4.1 cells, as evidenced by sub-G(1) cells and annexin V binding assay. This apoptotic process was accompanied by caspase-3 activation and PARP cleavage. Although caspase-3 inhibitor significantly reduced the activity of caspase-3, all of the caspase inhibitors tested in this study failed to rescue As4.1 cells from FCCP-induced cell death. In conclusion, this study demonstrated that FCCP, as a mitochondria-damaging agent, potently induces apoptosis in As4.1 juxtaglomerular cells via a caspase-independent manner.


Subject(s)
Apoptosis/drug effects , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Juxtaglomerular Apparatus/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/physiology , Caspase 3/metabolism , Caspase Inhibitors , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Juxtaglomerular Apparatus/cytology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/physiology , Uncoupling Agents/pharmacology
17.
Drug Chem Toxicol ; 33(4): 367-76, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20545600

ABSTRACT

MG132, as a proteasome inhibitor, has been shown to induce apoptotic cell death through the formation of reactive oxygen species (ROS). In this study, we investigated the effects of MG132 and/or MAPK inhibitors on As4.1 juxtaglomerular cells in relation to cell growth, cell death, ROS, and glutathione (GSH) levels. MG132 inhibited the growth of As4.1 cells and induced cell death, accompanied by the loss of mitochondrial membrane potential (MMP; DeltaPsi(m)) and activation of caspase-3 and -8. MG132 increased ROS levels, and GSH depleted cell numbers. The MEK inhibitor slightly reduced cell growth and caspase-3 activity in MG132-treated As4.1 cells and mildly increased MMP (DeltaPsi(m)) loss and O(2)(*-) level. However, it did not increase apoptosis and GSH depletion. The JNK inhibitor did not strongly influence cell growth, cell death, and GSH depletion by MG132, but increased caspase-3 activity, MMP (DeltaPsi(m)) loss, and O(2)(*-) level. Treatment with the p38 inhibitor magnified cell-growth inhibition and apoptosis by MG132. This agent also strongly increased caspase-8 activity, MMP (DeltaPsi(m)) loss, O(2)(*-) level, and GSH depletion. Conclusively, the p38 inhibitor strongly intensified cell death in MG132-treated As4.1 cells. The changes of GSH content by MG132 and/or MAPK inhibitors were closely related to the death of As4.1 cells.


Subject(s)
Apoptosis/drug effects , Glutathione/metabolism , Juxtaglomerular Apparatus/drug effects , Leupeptins/pharmacology , Proteasome Inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Caspase 3/metabolism , Caspase 8/metabolism , Cell Death/drug effects , Cell Line, Tumor , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/enzymology , Juxtaglomerular Apparatus/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Reactive Oxygen Species/metabolism
18.
J Am Soc Nephrol ; 21(6): 986-92, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20395378

ABSTRACT

Angiotensin II converting enzyme inhibitors (ACEI) or angiotensin II receptor blockers (ARB) presumably stimulate renin secretion by interrupting angiotensin II feedback inhibition. The increase in cytosolic calcium caused by activation of Gq-coupled AT1 receptors may mediate the renin-inhibitory effect of angiotensin II at the cellular level, implying that ACEI and ARB may work by reducing intracellular calcium. Here, we investigated whether angiotensin II blockade acts predominantly through Gs-mediated stimulation of adenylyl cyclase (AC) by testing the effect of ACEI and ARB in mice with juxtaglomerular cell-specific deficiency of the AC-stimulatory Gsalpha. The ACEI captopril and quinaprilate and the ARB candesartan significantly increased plasma renin concentration (PRC) to 20 to 40 times basal PRC in wild-type mice but did not significantly alter PRC in Gsalpha-deficient mice. Captopril also completely abrogated renin stimulation in wild-type mice after co-administration of propranolol, indomethacin, and L-NAME. Treatment with enalapril and a low-NaCl diet for 7 days led to a 35-fold increase in PRC among wild-type mice but no significant change in PRC among Gsalpha-deficient mice. Three different pharmacologic inhibitors of AC reduced the stimulatory effect of captopril by 70% to 80%. In conclusion, blockade of angiotensin II stimulates renin synthesis and release indirectly through the action of ligands that activate the cAMP/PKA pathway in a Gsalpha-dependent fashion, including catecholamines, prostaglandins, and nitric oxide.


Subject(s)
Adenylyl Cyclases/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II/antagonists & inhibitors , Angiotensin-Converting Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, Gs/metabolism , Juxtaglomerular Apparatus/metabolism , Renin/metabolism , Angiotensin II/metabolism , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Captopril/pharmacology , Catecholamines/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enalapril/pharmacology , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Juxtaglomerular Apparatus/drug effects , Male , Mice , Mice, Knockout , Models, Animal , Nitric Oxide/metabolism , Prostaglandins/metabolism , Signal Transduction/drug effects , Tetrahydroisoquinolines/pharmacology , Tetrazoles/pharmacology
19.
Arch Toxicol ; 84(8): 631-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20191265

ABSTRACT

Pyrogallol (PG) induces apoptosis in several types of cells mediated by superoxide anion (O(2*-)). Here, we investigated the effects of PG and/or MAPK (MEK, JNK, and p38) inhibitors on the changes in cell growth, cell death, reactive oxygen species (ROS), and GSH levels in As4.1 juxtaglomerular (JG) cells. PG inhibited the growth of As4.1 cells. It also induced apoptosis and the loss of mitochondrial membrane potential (MMP; DeltaPsi(m)) and increased the level of p53 protein. Intracellular O2(*-) level was increased in PG-treated As4.1 cells. PG also increased the number of GSH deleted cells in As4.1 cells. All the MAPK inhibitors significantly attenuated the growth inhibition and death mediated by PG. They decreased the levels of p53 protein and MMP (DeltaPsi(m)) loss in PG-treated As4.1 cells. They also reduced O2(*-) level and GSH-depleted cell number in these cells. In conclusion, MAPK inhibitors attenuated As4.1 cell growth inhibition and death mediated by PG treatment. The changes in O2(*-) and GSH levels by PG and/or MAPK inhibitors appeared to affect the growth and death of As4.1 cells.


Subject(s)
Glutathione/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Oxidants/toxicity , Protective Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrogallol/toxicity , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Humans , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/drug effects , Juxtaglomerular Apparatus/metabolism , Reactive Oxygen Species/metabolism
20.
Anticancer Res ; 29(11): 4423-31, 2009 Nov.
Article in English | MEDLINE | ID: mdl-20032388

ABSTRACT

Antimycin A (AMA) inhibits succinate oxidase, NADH oxidase and mitochondrial electron transport chain between cytochrome b and c. Here, we investigated the effects of AMA and/or mitogen-activated protein kinase (MAPK) inhibitors on As4.1 juxtaglomerular cells in relation to cell growth, cell death, reactive oxygen species (ROS) and glutathione (GSH) levels. Treatment with 50 nM AMA inhibited the growth of As4.1 cells at 48 hours and induced apoptosis, which was accompanied by the loss of mitochondrial membrane potential (DeltaPsi(m)). AMA increased ROS levels including that of intracellular O(2)(*-). AMA also induced GSH depletion. MEK inhibitor did not affect cell growth, cell death, DeltaPsi(m) loss, ROS level or GSH depletion in AMA-treated As4.1 cells. c-Jun N-terminal kinase (JNK) inhibitor also did not influence cell growth, cell death, ROS level and GSH depletion but did slightly increase DeltaPsi(m) loss. Treatment with p38 inhibitor magnified cell growth inhibition by AMA and increased cell death, DeltaPsi(m) loss and GSH depletion in AMA-treated As4.1 cells. Conclusively, p38 inhibitor intensified cell death in AMA-treated As4.1 cells. The changes of GSH content rather than ROS level by AMA and/or MAPK inhibitors were more closely related to the growth and death of As4.1 cells.


Subject(s)
Antimycin A/pharmacology , Glutathione/metabolism , Juxtaglomerular Apparatus/drug effects , Protein Kinase Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Cycle/drug effects , Cell Death/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Flavonoids/pharmacology , Imidazoles/pharmacology , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/enzymology , Juxtaglomerular Apparatus/metabolism , Kidney Neoplasms , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...