Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jeanette M.G. van Gool is active.

Publication


Featured researches published by Jeanette M.G. van Gool.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2008

Aliskiren-Binding Increases the Half Life of Renin and Prorenin in Rat Aortic Vascular Smooth Muscle Cells

Wendy W. Batenburg; René J.A. de Bruin; Jeanette M.G. van Gool; Dominik Müller; Michael Bader; Genevieve Nguyen; A.H. Jan Danser

Objective—Renin inhibition with aliskiren has been reported to cause a greater rise in renin than other types of renin-angiotensin system blockade, thereby potentially leading to angiotensin generation or stimulation of the human (pro)renin receptor (h(P)RR). Here we studied whether this rise in renin is attributable to an aliskiren-induced change in the prorenin conformation, allowing its detection in renin assays, or a change in renin/prorenin clearance. We also investigated whether aliskiren affects (pro)renin binding to its receptors, using rat aortic vascular smooth muscle cells (VSMCs) overexpressing the h(P)RR. Methods and Results—A 48-hour incubation with aliskiren at 4°C converted the prorenin conformation from “closed” to “open,” thus allowing its recognition in active site-directed renin assays. VSMCs accumulated (pro)renin through binding to mannose 6-phosphate receptors (M6PRs) and h(P)RRs. Aliskiren did not affect binding at 4°C. At 37°C, aliskiren increased (pro)renin accumulation up to 40-fold, and M6PR blockade prevented this. Aliskiren increased the intracellular half life of prorenin 2 to 3 times. Conclusion—Aliskiren allows the detection of prorenin as renin, and decreases renin/prorenin clearance. Both phenomena may contribute to the “renin” surge during aliskiren treatment, but because they depend on aliskiren binding, they will not result in angiotensin generation. Aliskiren does not affect (pro)renin binding to its receptors.


Hypertension | 2008

Aliskiren Accumulates in Renin Secretory Granules and Binds Plasma Prorenin

Manne Krop; Ingrid M. Garrelds; René J.A. de Bruin; Jeanette M.G. van Gool; Naomi D.L. Fisher; Norman K. Hollenberg; A.H. Jan Danser

The vascular effects of aliskiren last longer than expected based on its half life, and this renin inhibitor has been reported to cause a greater renin rise than other renin-angiotensin system blockers. To investigate whether aliskiren accumulation in secretory granules contributes to these phenomena, renin-synthesizing mast cells were incubated with aliskiren, washed, and exposed to forskolin in medium without aliskiren (0.1 to 1000 nmol/L). (Pro)renin concentrations were measured by renin- and prorenin-specific immunoradiometric assays, and renin activity was measured by enzyme-kinetic assay. Without aliskiren, the culture medium predominantly contained prorenin, the cells exclusively stored renin, and forskolin doubled renin release. Aliskiren dose-dependently bound to (pro)renin in the medium and cell lysates and did not alter the effect of forskolin. The aliskiren concentrations required to bind prorenin were 1 to 2 orders of magnitude higher than those needed to bind renin. Blockade of cell lysate renin activity ranged from 27±15% to 79±5%, and these percentages were identical for the renin that was released by forskolin, indicating that they represented the same renin pool, ie, the renin storage granules. Comparison of renin and prorenin measurements in blood samples obtained from human volunteers treated with aliskiren, both before and after prorenin activation, revealed that ≤30% of prorenin was detected in renin-specific assays. In conclusion, aliskiren accumulates in renin granules, thus allowing long-lasting renin-angiotensin system blockade beyond the half-life of this drug. Aliskiren also binds to prorenin. This allows its detection as renin, and might explain, in part, the renin rise during renin inhibition.


Hypertension | 2001

Angiotensin II Type 2 Receptors and Cardiac Hypertrophy in Women With Hypertrophic Cardiomyopathy

Jaap Deinum; Jeanette M.G. van Gool; Marcel Kofflard; Folkert J. ten Cate; A.H. Jan Danser

The development of left ventricular hypertrophy in subjects with hypertrophic cardiomyopathy (HCM) is variable, suggesting a role for modifying factors such as angiotensin II. Angiotensin II mediates both trophic and antitrophic effects, via angiotensin II type 1 (AT1-R) and angiotensin II type 2 (AT2-R) receptors, respectively. Here we investigated the effect of the AT2-R gene A/C3123 polymorphism, located in the 3′ untranslated region of exon 3, on left ventricular mass index (LVMI) in 103 genetically independent subjects with HCM (age, 12 to 81 years). LVMI and interventricular septum thickness were determined by 2D echocardiography. Extent of hypertrophy was quantified by a point score (Wigle score). Plasma prorenin, renin, and ACE were determined by immunoradiometric or fluorometric assays, and genotyping was performed by polymerase chain reaction. In men, no associations between AT2-R genotype and any of the measured parameters were observed, whereas in women, LVMI decreased with the number of C alleles (211±19, 201±18, and 152±10 g/m2 in women with the AA, AC, and CC genotype, respectively;P =0.015). Similar C allele–related decreases in women were observed for interventricular septum thickness (P =0.13), Wigle score (P =0.05), plasma renin (P =0.03), and plasma prorenin (P =0.26). Multiple regression analysis revealed that the AT2-R C allele–related effect on LVMI (&bgr;=−30.7±11.1, P =0.010) occurred independently of plasma renin, the AT1-R gene A/C1166 polymorphism, or the ACE gene I/D polymorphism. In conclusion, AT2-Rs modulate cardiac hypertrophy in women with HCM, independently of the circulating renin-angiotensin system. These data support the contention that AT2-Rs mediate antitrophic effects in humans.


Journal of Hypertension | 2011

Urinary renin, but not angiotensinogen or aldosterone, reflects the renal renin-angiotensin-aldosterone system activity and the efficacy of renin-angiotensin-aldosterone system blockade in the kidney.

Mieke van den Heuvel; Wendy W. Batenburg; Sjaam Jainandunsing; Ingrid M. Garrelds; Jeanette M.G. van Gool; Richard A. Feelders; Anton H. van den Meiracker; A.H. Jan Danser

Objective To study which renin–angiotensin–aldosterone system (RAAS) component best reflects renal RAAS activity. Methods and results We measured urinary and plasma renin, prorenin, angiotensinogen, aldosterone, albumin and creatinine in 101 diabetic and nondiabetic patients with or without hypertension. Plasma prorenin was elevated in diabetic patients. Urinary prorenin was undetectable. Urinary albumin and renin were higher in diabetic patients. Men had higher plasma renin/prorenin levels, and lower plasma angiotensinogen levels than women. Plasma creatinine and albumin were also higher in men. Urinary RAAS components showed no sexual dimorphism, whereas urinary creatinine and albumin were higher in men. Angiotensin-converting enzyme inhibitors and angiotensin II type 1 receptor blockers increased plasma renin and decreased plasma angiotensinogen, without altering plasma aldosterone. In contrast, in urine, these drugs decreased renin and aldosterone without affecting angiotensinogen. When analyzing all patients together, urinary angiotensinogen excretion closely mimicked that of albumin, whereas urinary angiotensinogen and albumin levels both were 0.05% or less of their concomitant plasma levels. This may reflect the identical glomerular filtration and tubular handling of both proteins, which have a comparable molecular weight. In contrast, urinary renin excretion did not correlate with urinary albumin excretion, and the urinary/plasma concentration ratio of renin was more than 200 times the ratio of albumin, despite its comparable molecular weight. Urinary aldosterone excretion closely followed urinary creatinine excretion. Conclusion The increased urinary renin levels in diabetes and the decreased urinary renin levels following RAAS blockade, occurring independently of changes in plasma renin, reflect the activated renal RAAS in diabetes and the success of RAAS blockade in the kidney, respectively. Urinary renin, therefore, more closely reflects renal RAAS activity than urinary angiotensinogen or aldosterone.


Hypertension | 2005

Selective Angiotensin-Converting Enzyme C-Domain Inhibition Is Sufficient to Prevent Angiotensin I–Induced Vasoconstriction

Joep H.M. van Esch; Beril Tom; Vincent Dive; Wendy W. Batenburg; Dimitris Georgiadis; Athanasios Yiotakis; Jeanette M.G. van Gool; René J.A. de Bruijn; René de Vries; A.H. Jan Danser

Somatic angiotensin-converting enzyme (ACE) contains 2 domains (C-domain and N-domain) capable of hydrolyzing angiotensin I (Ang I) and bradykinin. Here we investigated the effect of the selective C-domain and N-domain inhibitors RXPA380 and RXP407 on Ang I–induced vasoconstriction of porcine femoral arteries (PFAs) and bradykinin-induced vasodilation of preconstricted porcine coronary microarteries (PCMAs). Ang I concentration-dependently constricted PFAs. RXPA380, at concentrations >1 &mgr;mol/L, shifted the Ang I concentration-response curve (CRC) 10-fold to the right. This was comparable to the maximal shift observed with the ACE inhibitors (ACEi) quinaprilat and captopril. RXP407 did not affect Ang I at concentrations ≤0.1 mmol/L. Bradykinin concentration-dependently relaxed PCMAs. RXPA380 (10 &mgr;mol/L) and RXP407 (0.1 mmol/L) potentiated bradykinin, both inducing a leftward shift of the bradykinin CRC that equaled ≈50% of the maximal shift observed with quinaprilat. Ang I added to blood plasma disappeared with a half life (t1/2) of 42±3 minutes. Quinaprilat increased the t1/2 ≈4-fold, indicating that 71±6% of Ang I metabolism was attributable to ACE. RXPA380 (10 &mgr;mol/L) and RXP407 (0.1 mmol/L) increased the t1/2 ≈2-fold, thereby suggesting that both domains contribute to conversion in plasma. In conclusion, tissue Ang I–II conversion depends exclusively on the ACE C-domain, whereas both domains contribute to conversion by soluble ACE and to bradykinin degradation at tissue sites. Because tissue ACE (and not plasma ACE) determines the hypertensive effects of Ang I, these data not only explain why N-domain inhibition does not affect Ang I–induced vasoconstriction in vivo but also why ACEi exert blood pressure–independent effects at low (C-domain–blocking) doses.


Hypertension | 2014

Neuron-Specific (Pro)renin Receptor Knockout Prevents the Development of Salt-Sensitive Hypertension

Wencheng Li; Hua Peng; Eamonn P. Mehaffey; Christie D. Kimball; Justin L. Grobe; Jeanette M.G. van Gool; Michelle N. Sullivan; Scott Earley; A.H. Jan Danser; Atsuhiro Ichihara; Yumei Feng

The (pro)renin receptor (PRR), which binds both renin and prorenin, is a newly discovered component of the renin–angiotensin system that is highly expressed in the central nervous system. The significance of brain PRRs in mediating local angiotensin II formation and regulating blood pressure remains unclear. The current study was performed to test the hypothesis that PRR-mediated, nonproteolytic activation of prorenin is the main source of angiotensin II in the brain. Thus, PRR knockout in the brain is expected to prevent angiotensin II formation and development of deoxycorticosterone acetate-salt–induced hypertension. A neuron-specific PRR (ATP6AP2) knockout mouse model was generated using the Cre-LoxP system. Physiological parameters were recorded by telemetry. PRR expression, detected by immunostaining and reverse transcription–polymerase chain reaction, was significantly decreased in the brains of knockout mice compared with wild-type mice. Intracerebroventricular infusion of mouse prorenin increased blood pressure and angiotensin II formation in wild-type mice. This hypertensive response was abolished in PRR-knockout mice in association with a reduction in angiotensin II levels. Deoxycorticosterone acetate-salt increased PRR expression and angiotensin II formation in the brains of wild-type mice, an effect that was attenuated in PRR-knockout mice. PRR knockout in neurons prevented the development of deoxycorticosterone acetate-salt–induced hypertension as well as activation of cardiac and vasomotor sympathetic tone. In conclusion, nonproteolytic activation of prorenin through binding to the PRR mediates angiotensin II formation in the brain. Neuron-specific PRR knockout prevents the development of deoxycorticosterone acetate-salt–induced hypertension, possibly through diminished angiotensin II formation.


Hypertension | 2009

Cardiac Renin Levels Are Not Influenced by the Amount of Resident Mast Cells

Manne Krop; Richard van Veghel; Ingrid M. Garrelds; René J.A. de Bruin; Jeanette M.G. van Gool; Anton H. van den Meiracker; Marco Thio; Paul L.A. van Daele; A.H. Jan Danser

To investigate whether mast cells release renin in the heart, we studied renin and prorenin synthesis by such cells, using the human mast cell lines human mastocytoma 1 and LAD2, as well as fresh mast cells from mastocytosis patients. We also quantified the contribution of mast cells to cardiac renin levels in control and infarcted rat hearts. Human mastocytoma 1 cells contained and released angiotensin I–generating activity, and the inhibition of this activity by the renin inhibitor aliskiren was comparable to that of recombinant human renin. Prorenin activation with trypsin increased angiotensin I–generating activity in the medium only, suggesting release but not storage of prorenin. The adenylyl cyclase activator forskolin, the cAMP analogue 8-db-cAMP, and the degranulator compound 48/80 increased renin release without affecting prorenin. Angiotensin II blocked the forskolin-induced renin release. Angiotensin I–generating activity was undetectable in LAD2 cells and fresh mast cells. Nonperfused rat hearts contained angiotensin I–generating activity, and aliskiren blocked ≈70% of this activity. A 30-minute buffer perfusion washed away >70% of the aliskiren-inhibitable angiotensin I–generating activity. Prolonged buffer perfusion or compound 48/80 did not decrease cardiac angiotensin I–generating activity further or induce angiotensin I–generating activity release in the perfusion buffer. Results in infarcted hearts were identical, despite the increased mast cell number in such hearts. In conclusion, human mastocytoma 1 cells release renin and prorenin, and the regulation of this release resembles that of renal renin. However, this is not a uniform property of all mast cells. Mast cells appear an unlikely source of renin in the heart, both under normal and pathophysiological conditions.


Hypertension | 2013

New Renin Inhibitor VTP-27999 Alters Renin Immunoreactivity and Does Not Unfold Prorenin

Manne Krop; Xifeng Lu; Koen Verdonk; Schalekamp Ma; Jeanette M.G. van Gool; Brian M. McKeever; Richard Gregg; A.H. Jan Danser

Renin inhibitors like aliskiren not only block renin but also bind prorenin, thereby inducing a conformational change (like the change induced by acid) allowing its recognition in a renin-specific assay. Consequently, aliskiren can be used to measure prorenin. VTP-27999 is a new renin inhibitor with an aliskiren-like IC50 and t1/2, and a much higher bioavailability. This study addressed (pro)renin changes during treatment of volunteers with VTP-27999 or aliskiren. Both drugs increased renin immunoreactivity. Treatment of plasma samples from aliskiren-treated subjects with excess aliskiren yielded higher renin immunoreactivity levels, confirming the presence of prorenin. Unexpectedly, this approach did not work in VTP-27999–treated subjects, although an assay detecting the prosegment revealed that their blood still contained prorenin. Subsequent in vitro analysis showed that VTP-27999 increased renin immunoreactivity for a given amount of renin by ≥30% but did not unfold prorenin. Yet, it did bind to acid-activated, intact prorenin and then again increased immunoreactivity in a renin assay. However, no such increase in immunoreactivity was seen when measuring acid-activated prorenin bound to VTP-27999 with a prosegment-directed assay. The VTP-27999–induced rises in renin immunoreactivity could be competitively prevented by aliskiren, and antibody displacement studies revealed a higher affinity of the active site-directed antibodies in the presence of VTP-27999. In conclusion, VTP-27999 increases renin immunoreactivity in renin immunoassays because it affects the affinity of the active site-directed antibody. Combined with its lack of effect on prorenin, these data show that VTP-27999 differs from aliskiren. The clinical relevance of these results needs to be established.


Hypertension | 2016

On the Origin of Urinary Renin: A Translational Approach.

Lodi C.W. Roksnoer; Bart F.J. Heijnen; Daisuke Nakano; Janos Peti-Peterdi; Stephen B. Walsh; Ingrid M. Garrelds; Jeanette M.G. van Gool; Robert Zietse; Harry A.J. Struijker-Boudier; Ewout J. Hoorn; A.H. Jan Danser

Urinary angiotensinogen excretion parallels albumin excretion, which is not the case for renin, while renin’s precursor, prorenin, is undetectable in urine. We hypothesized that renin and prorenin, given their smaller size, are filtered through the glomerulus in larger amounts than albumin and angiotensinogen, and that differences in excretion rate are because of a difference in reabsorption in the proximal tubule. To address this, we determined the glomerular sieving coefficient of renin and prorenin and measured urinary renin/prorenin 1) after inducing prorenin in Cyp1a1-Ren2 rats and 2) in patients with Dent disease or Lowe syndrome, disorders characterized by defective proximal tubular reabsorption. Glomerular sieving coefficients followed molecular size (renin>prorenin>albumin). The induction of prorenin in rats resulted in a >300-fold increase in plasma prorenin and doubling of blood pressure but did not lead to the appearance of prorenin in urine. It did cause parallel rises in urinary renin and albumin, which losartan but not hydralazine prevented. Defective proximal tubular reabsorption increased urinary renin and albumin 20- to 40-fold, and allowed prorenin detection in urine, at ≈50% of its levels in plasma. Taken together, these data indicate that circulating renin and prorenin are filtered into urine in larger amounts than albumin. All 3 proteins are subsequently reabsorbed in the proximal tubule. For prorenin, such reabsorption is ≈100%. Minimal variation in tubular reabsorption (in the order of a few %) is sufficient to explain why urinary renin and albumin excretion do not correlate. Urinary renin does not reflect prorenin that is converted to renin in tubular fluid.Urinary angiotensinogen excretion parallels albumin excretion, which is not the case for renin, while renin’s precursor, prorenin, is undetectable in urine. We hypothesized that renin and prorenin, given their smaller size, are filtered through the glomerulus in larger amounts than albumin and angiotensinogen, and that differences in excretion rate are because of a difference in reabsorption in the proximal tubule. To address this, we determined the glomerular sieving coefficient of renin and prorenin and measured urinary renin/prorenin 1) after inducing prorenin in Cyp1a1-Ren2 rats and 2) in patients with Dent disease or Lowe syndrome, disorders characterized by defective proximal tubular reabsorption. Glomerular sieving coefficients followed molecular size (renin>prorenin>albumin). The induction of prorenin in rats resulted in a >300-fold increase in plasma prorenin and doubling of blood pressure but did not lead to the appearance of prorenin in urine. It did cause parallel rises in urinary renin and albumin, which losartan but not hydralazine prevented. Defective proximal tubular reabsorption increased urinary renin and albumin 20- to 40-fold, and allowed prorenin detection in urine, at ≈50% of its levels in plasma. Taken together, these data indicate that circulating renin and prorenin are filtered into urine in larger amounts than albumin. All 3 proteins are subsequently reabsorbed in the proximal tubule. For prorenin, such reabsorption is ≈100%. Minimal variation in tubular reabsorption (in the order of a few %) is sufficient to explain why urinary renin and albumin excretion do not correlate. Urinary renin does not reflect prorenin that is converted to renin in tubular fluid.


Journal of Hypertension | 2011

Evaluation of a direct prorenin assay making use of a monoclonal antibody directed against residues 32-39 of the prosegment.

Manne Krop; Jeanette M.G. van Gool; Duane Day; Norman K. Hollenberg; Alexander H. Jan Danser

Background Prorenin is an early marker of microvascular complications in diabetes. However, it can only be measured indirectly (following its conversion to renin), with a renin immunoradiometric assay (IRMA). Unfortunately, treatment with a renin inhibitor interferes with this assay, because renin inhibitors induce a conformational change in prorenin, thereby allowing its detection as renin. Methods We evaluated Molecular Innovations new direct prorenin ELISA, which makes use of an antibody that recognizes an epitope near prorenins putative cleavage site (R43L44), thus no longer requiring prorenin activation. Plasma samples of 41 diabetic individuals treated with aliskiren (renin inhibitor) or irbesartan were tested. Semi-purified recombinant prorenin was used as standard, because the ELISA standard yielded approximately 10-fold lower values in the renin IRMA following its conversion to renin. Results The ELISA detected prorenin levels that were identical to those determined by the IRMA in untreated and irbesartan-treated individuals. Yet, it yielded higher prorenin levels in aliskiren-treated individuals. Aliskiren, at levels reached in plasma during treatment, did not interfere with the ELISA, but allowed the detection of up to 20–30% of prorenin as renin in the IRMA, thereby resulting in a significant overestimation of renin and an underestimation of prorenin. The ELISA rendered results within 2 h and did not require a pretreatment period of several days to convert prorenin to renin. Conclusion The new direct assay allows rapid prorenin detection, is not hampered by aliskiren when used at clinically relevant doses, and might be used to identify diabetic patients developing retinopathy and/or nephropathy.

Collaboration


Dive into the Jeanette M.G. van Gool's collaboration.

Top Co-Authors

Avatar

A.H. Jan Danser

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Ingrid M. Garrelds

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Ewout J. Hoorn

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Lodi C.W. Roksnoer

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Manne Krop

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Robert Zietse

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wendy W. Batenburg

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Janos Peti-Peterdi

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge