Network


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

Hotspot


Dive into the research topics where Kirsten Madsen is active.

Publication


Featured researches published by Kirsten Madsen.


Journal of The American Society of Nephrology | 2010

Angiotensin II Promotes Development of the Renal Microcirculation through AT1 Receptors

Kirsten Madsen; Niels Marcussen; Michael Pedersen; Gitte Kjærsgaard; Carie S. Facemire; Thomas M. Coffman; Boye L. Jensen

Pharmacologic or genetic deletion of components of the renin-angiotensin system leads to postnatal kidney injury, but the roles of these components in kidney development are unknown. To test the hypothesis that angiotensin II supports angiogenesis during postnatal kidney development, we quantified CD31(+) postglomerular microvessels, performed quantitative PCR analysis of vascular growth factor expression, and measured renal blood flow by magnetic resonance. Treating rats with the angiotensin II type 1 receptor antagonist candesartan for 2 weeks after birth reduced the total length, volume, and surface area of capillaries in both the cortex and the medulla and inhibited the organization of vasa recta bundles. In addition, angiotensin II type 1 antagonism inhibited the transcription of angiogenic growth factors vascular endothelial growth factor, angiopoietin-1, angiopoietin-2, and the angiopoietin receptor Tie-2 in cortex and medulla. Similarly, Agtr1a(-/-);Agtr1b(-/-) mouse kidneys had decreased angiopoietin-1, angiopoietin-2, and Tie-2 mRNAs at postnatal day 14. To test whether increased urinary flow leads to microvascular injury, we induced postnatal polyuria with either lithium or adrenalectomy, but these did not alter vascular endothelial growth factor expression or vasa recta organization. Compared with vehicle-treated rats, renal blood flow was significantly (approximately 20%) lower in candesartan-treated rats even 14 days after candesartan withdrawal. Taken together, these data demonstrate that angiotensin II promotes postnatal expansion of postglomerular capillaries and organization of vasa recta bundles, which are necessary for development of normal renal blood flow.


Nephron Physiology | 2010

High-Level Connexin Expression in the Human Juxtaglomerular Apparatus

Lisa Kurtz; Kirsten Madsen; Birgül Kurt; Boye L. Jensen; Steen Walter; Bernhard Banas; Charlotte Wagner; Armin Kurtz

Recent evidence obtained in rodents indicates that gap junctions in the juxtaglomerular apparatus play an important role in the control of renin-producing cells and in tubuloglomerular signaling. These gap junctions are formed by cell-specific expression patterns of the vascular connexins Cx37, Cx40, Cx43 and Cx45. In order to obtain a first indication if gap junctions might play a similar important functional role in the juxtaglomerular apparatus of human kidneys, this study aimed to characterize the juxtaglomerular localization of Cx40, Cx37, Cx43 and Cx45 in human kidney specimens. We found Cx37, Cx40 and Cx43, but not Cx45 expression in high density in the extraglomerular mesangium. Renin-producing cells displayed strong immunoreactivity for Cx40 and Cx37. Cx37, Cx40 and Cx43 were also seen in the endothelium of arteries/arterioles outside of the glomeruli, whereas Cx45 was located in vascular smooth muscle cells. All four connexins were also expressed within the glomeruli. These findings indicate that the expression pattern of vascular connexins in the human kidney cortex is very similar to that previously found for mouse and rat kidneys, suggesting that the intrarenal expression pattern of vascular connexins is conserved among the mammalian species. Because of this similarity, and in particular in view of the strong expression of Cx37 and Cx40 in the juxtaglomerular area, we infer that those functions of connexins that have already been demonstrated for rodent kidneys, such as a central role of Cx40 for the development and function of renin-producing cells and for tubuloglomerular signal transmission, might hold for human kidneys as well.


Pflügers Archiv: European Journal of Physiology | 2013

Regulation of renin secretion by renal juxtaglomerular cells

Ulla G. Friis; Kirsten Madsen; Jane Stubbe; Pernille B. Lærkegaard Hansen; Per Svenningsen; Peter Bie; Ole Skøtt; Boye L. Jensen

A major rate-limiting step in the renin–angiotensin–aldosterone system is the release of active renin from endocrine cells (juxtaglomerular (JG) cells) in the media layer of the afferent glomerular arterioles. The number and distribution of JG cells vary with age and the physiological level of stimulation; fetal life and chronic stimulation by extracellular volume contraction is associated with recruitment of renin-producing cells. Upon stimulation of renin release, labeled renin granules “disappear;” the number of granules decrease; cell membrane surface area increases in single cells, and release is quantal. Together, this indicates exocytosis as the predominant mode of release. JG cells release few percent of total renin content by physiological stimulation, and recruitment of renin cells is preferred to recruitment of granules during prolonged stimulation. Several endocrine and paracrine agonists, neurotransmitters, and cell swelling converge on the stimulatory cyclic AMP (cAMP) pathway. Renin secretion is attenuated in mice deficient in beta-adrenoceptors, prostaglandin E2–EP4 receptors, Gsα protein, and adenylyl cyclases 5 and 6. Phosphodiesterases (PDE) 3 and 4 degrade cAMP in JG cells, and PDE3 is inhibited by cyclic GMP (cGMP) and couples the cGMP pathway to the cAMP pathway. Cyclic AMP enhances K+-current in JG cells and is permissive for secretion by stabilizing membrane potential far from threshold that activates L-type voltage-gated calcium channels. Intracellular calcium paradoxically inhibits renin secretion likely through attenuated formation and enhanced degradation of cAMP; by activation of chloride currents and interaction with calcineurin. Connexin 40 is necessary for localization of JG cells in the vascular wall and for pressure- and macula densa-dependent suppression of renin release.


Hypertension | 2011

Stimulation of Renin Secretion by Catecholamines Is Dependent on Adenylyl Cyclases 5 and 6

F. Aldehni; T. Tang; Kirsten Madsen; M. Plattner; A. Schreiber; Ulla G. Friis; H. K. Hammond; P. L. Han; Frank Schweda

The sympathetic nervous system stimulates renin release from juxtaglomerular cells via the &bgr;-adrenoreceptor-cAMP pathway. Recent in vitro studies have suggested that the calcium-inhibited adenylyl cyclases (ACs) 5 and 6 possess key roles in the control of renin exocytosis. To investigate the relative contribution of AC5 and AC6 to the regulation of renin release in vivo we performed experiments using AC5 and AC6 knockout mice. Male AC5−/− mice exhibited normal plasma renin concentrations, renal renin synthesis (mRNA and renin content), urinary volume, and systolic blood pressure. In male AC6−/− mice, plasma renin concentration (AC6−/−: 732±119; AC6 +/+: 436±78 ng of angiotensin I per hour*mL−1; P<0.05), and renin synthesis were stimulated associated with an increased excretion of dilute urine (1.55-fold; P<0.05) and reduced blood pressure (−10.6 mm Hg; P<0.001). Stimulation of plasma renin concentration by a single injection of the &bgr;-adrenoreceptor agonist isoproterenol (10 mg/kg IP) was significantly attenuated in AC5−/− (male: −20%; female: −33%) compared with wild-type mice in vivo. The mitigation of the plasma renin concentration response to isoproterenol was even more pronounced in AC6−/− (male: −63%; female: −50% versus AC6+/+). Similarly, the effects of isoproterenol, prostaglandin E2, and pituitary adenylyl cyclase-activating polypeptide on renin release from isolated perfused kidneys were attenuated to a higher extent in AC6−/− (−51% to −98% versus AC6+/+) than in AC5−/− (−31% to 46% versus AC5+/+). In conclusion, both AC5 and AC6 are involved in the stimulation of renin secretion in vivo, and AC6 is the dominant isoforms in this process.


American Journal of Physiology-renal Physiology | 2012

Tissue injury after lithium treatment in human and rat postnatal kidney involves glycogen synthase kinase-3β-positive epithelium.

Gitte Kjærsgaard; Kirsten Madsen; Niels Marcussen; Sten Christensen; Steen Walter; Boye L. Jensen

It was hypothesized that lithium causes accelerated and permanent injury to the postnatally developing kidney through entry into epithelial cells of the distal nephron and inhibition of glycogen synthase kinase-3β (GSK-3β). GSK-3β immunoreactivity was associated with glomeruli, the thick ascending limb of Henles loop, and collecting ducts in the developing and adult human and rat kidney. In rats, the abundance of inactive, phosphorylated GSK-3β (pGSK-3β) protein decreased during postnatal development. After feeding of dams with litters lithium [50 mmol Li/kg chow, postnatal (P) days 7-28], the offspring showed plasma lithium concentration of 1.0 mmol/l. Kidneys from lithium-treated rat pups exhibited dilated distal nephron segments with microcysts. Stereological analysis showed reduced cortex and outer medullary volumes. Lithium increased pGSK-3β and the proliferation marker proliferating cell nuclear antigen (PCNA) protein abundances in the cortex and medulla. After lithium treatment, pGSK-3β-immunopositive cells exhibited restricted distribution and were associated primarily with subsets of cells in dilated and microcystic segments of cortical collecting ducts. After 6 wk of lithium discontinuation, adult rats exhibited attenuated urine concentration capacity and diminished outer medullary volume. Histological sections of two nephrectomy samples and a biopsy from three long-term lithium-treated patients showed multiple cortical microcysts that originated from normally appearing tubules. Microcysts were lined by a cuboidal PCNA-, GSK-3β-, and pGSK-3β-immunopositive epithelium. The postnatal rat kidney may serve as an experimental model for the study of lithium-induced human kidney injury. The data are compatible with a causal relationship between epithelial entry of lithium into cells of the aldosterone-sensitive distal nephron, inactivation of GSK-3β, proliferation, and microcysts.


Circulation | 2013

Loss of Collectrin, an Angiotensin-Converting Enzyme 2 Homolog, Uncouples Endothelial Nitric Oxide Synthase and Causes Hypertension and Vascular Dysfunction

Sylvia Cechova; Qing Zeng; Marie Billaud; Stephanie Mutchler; Christine K. Rudy; Adam C. Straub; Liqun Chi; Fang R. Chan; Jun Hu; Robert I. Griffiths; Nancy L. Howell; Kirsten Madsen; Boye L. Jensen; Lisa A. Palmer; Robert M. Carey; Sun-Sang J. Sung; Sandra M. Malakauskas; Brant E. Isakson; Thu H. Le

Background— Collectrin is an orphan member of the renin-angiotensin system and is a homolog of angiotensin-converting enzyme 2, sharing ≈50% sequence identity. Unlike angiotensin-converting enzyme 2, collectrin lacks any catalytic domain. Collectrin has been shown to function as a chaperone of amino acid transporters. In rodents, the renal expression of collectrin is increased after subtotal nephrectomy and during high-salt feeding, raising the question of whether collectrin has any direct role in blood pressure regulation. Methods and Results— Using a susceptible genetic background, we demonstrate that deletion of collectrin results in hypertension, exaggerated salt sensitivity, and impaired pressure natriuresis. Collectrin knockout mice display impaired endothelium-dependent vasorelaxation that is associated with vascular remodeling, endothelial nitric oxide synthase uncoupling, decreased nitric oxide production, and increased superoxide generation. Treatment with Tempol, a superoxide scavenger, attenuates the augmented sodium sensitivity in collectrin knockout mice. We report for the first time that collectrin is expressed in endothelial cells. Furthermore, collectrin directly regulates L-arginine uptake and plasma membrane levels of CAT1 and y+LAT1 amino acid transporters in endothelial cells. Treatment with L-arginine modestly lowers blood pressure of collectrin knockout mice. Conclusions— Collectrin is a consequential link between the transport of L-arginine and endothelial nitric oxide synthase uncoupling in hypertension.


Circulation | 2013

Loss of Collectrin, an ACE2 Homologue, Uncouples Endothelial Nitric Oxide Synthase and Causes Hypertension and Vascular Dysfunction

Sylvia Cechova; Qing Zeng; Marie Billaud; Stephanie Mutchler; Christine K. Rudy; Adam C. Straub; Fang R. Chan; Liqun Chi; Jun Hu; Robert Griffiths; Nancy L. Howell; Kirsten Madsen; Boye L. Jensen; Lisa A. Palmer; Robert M. Carey; Sun-Sang J. Sung; Sandra Malakauskas; Brant E. Isakson; Thu H. Le

Background— Collectrin is an orphan member of the renin-angiotensin system and is a homolog of angiotensin-converting enzyme 2, sharing ≈50% sequence identity. Unlike angiotensin-converting enzyme 2, collectrin lacks any catalytic domain. Collectrin has been shown to function as a chaperone of amino acid transporters. In rodents, the renal expression of collectrin is increased after subtotal nephrectomy and during high-salt feeding, raising the question of whether collectrin has any direct role in blood pressure regulation. Methods and Results— Using a susceptible genetic background, we demonstrate that deletion of collectrin results in hypertension, exaggerated salt sensitivity, and impaired pressure natriuresis. Collectrin knockout mice display impaired endothelium-dependent vasorelaxation that is associated with vascular remodeling, endothelial nitric oxide synthase uncoupling, decreased nitric oxide production, and increased superoxide generation. Treatment with Tempol, a superoxide scavenger, attenuates the augmented sodium sensitivity in collectrin knockout mice. We report for the first time that collectrin is expressed in endothelial cells. Furthermore, collectrin directly regulates L-arginine uptake and plasma membrane levels of CAT1 and y+LAT1 amino acid transporters in endothelial cells. Treatment with L-arginine modestly lowers blood pressure of collectrin knockout mice. Conclusions— Collectrin is a consequential link between the transport of L-arginine and endothelial nitric oxide synthase uncoupling in hypertension.


Journal of The American Society of Nephrology | 2009

Hypotonicity-Induced Renin Exocytosis from Juxtaglomerular Cells Requires Aquaporin-1 and Cyclooxygenase-2

Ulla G. Friis; Kirsten Madsen; Per Svenningsen; Pernille B. Lærkegaard Hansen; Ambika Gulaveerasingam; Finn Jørgensen; Christian Aalkjaer; Ole Skøtt; Boye L. Jensen

The mechanism by which extracellular hypotonicity stimulates release of renin from juxtaglomerular (JG) cells is unknown. We hypothesized that osmotically induced renin release depends on water movement through aquaporin-1 (AQP1) water channels and subsequent prostanoid formation. We recorded membrane capacitance (C(m)) by whole-cell patch clamp in single JG cells as an index of exocytosis. Hypotonicity increased C(m) significantly and enhanced outward current. Indomethacin, PLA(2) inhibition, and an antagonist of prostaglandin transport impaired the C(m) and current responses to hypotonicity. Hypotonicity also increased exocytosis as determined by a decrease in single JG cell quinacrine fluorescence in an indomethacin-sensitive manner. In single JG cells from COX-2(-/ -) and AQP1(-/ -) mice, hypotonicity increased neither C(m) nor outward current, but 0.1-muM PGE(2) increased both in these cells. A reduction in osmolality enhanced cAMP accumulation in JG cells but not in renin-producing As4.1 cells; only the former had detectable AQP1 expression. Inhibition of protein kinase A blocked the hypotonicity-induced C(m) and current response in JG cells. Taken together, our results show that a 5 to 7% decrease in extracellular tonicity leads to AQP1-mediated water influx in JG cells, PLA(2)/COX-2-mediated prostaglandin-dependent formation of cAMP, and activation of PKA, which promotes exocytosis of renin.


Hypertension | 2010

Increased Renin Production in Mice With Deletion of Peroxisome Proliferator-Activated Receptor-γ in Juxtaglomerular Cells

Michael Desch; Andrea Schreiber; Frank Schweda; Kirsten Madsen; Ulla G. Friis; Eric T. Weatherford; Curt D. Sigmund; Maria Luisa S. Sequeira Lopez; R. Ariel Gomez; Vladimir T. Todorov

We recently found that endogenous (free fatty acids) and pharmacological (thiazolidinediones) agonists of nuclear receptor Peroxisome proliferator-activated receptor (PPAR)&ggr; stimulate renin transcription. In addition, the renin gene was identified as a direct target of PPAR&ggr;. The mouse renin gene is regulated by PPAR&ggr; through a distal enhancer direct repeat closely related to consensus PPAR response element (PPRE). In vitro studies demonstrated that PPAR&ggr; knockdown stimulated PPRE-driven transcription. These data predicted that deficiency of PPAR&ggr; would upregulate mouse renin expression. Consistent with these observations knockdown of PPAR&ggr; increased the transcription of a reporter gene driven by the mouse renin PPRE-like motif in vitro. To study the impact of PPAR&ggr; on renin production in vivo, we used a cre/lox system to generate double-transgenic mice with disrupted PPAR&ggr; locus in renin-producing juxtaglomerular (JG) cells of the kidney (RC-PPAR&ggr;fl/fl mice). We provide evidence that PPAR&ggr; expression was effectively reduced in JG cells of RC-PPAR&ggr;fl/fl mice. Fluorescent immunohistochemistry showed stronger renin signal in RC-PPAR&ggr;fl/fl than in littermate control RC-PPAR&ggr;wt/wt mice. Renin mRNA levels and plasma renin concentration in RC-PPAR&ggr;fl/fl mice were almost 2-fold higher than in littermate controls. Arterial blood pressure and pressure control of renal vascular resistance, which play decisive roles in the regulation of renin production were indistinguishable between RC-PPAR&ggr;wt/wt and RC-PPAR&ggr;fl/fl mice. These data demonstrate that the JG-specific PPAR&ggr; deficiency results in increased mouse renin expression in vivo thus corroborating earlier in vitro results. PPAR&ggr; appears to be a relevant transcription factor for the control of renin gene in JG cells.


Kidney International | 2010

Inhibition of calcineurin phosphatase promotes exocytosis of renin from juxtaglomerular cells.

Kirsten Madsen; Ulla G. Friis; Jennifer L. Gooch; Pernille B. Lærkegaard Hansen; Lisbeth Holmgaard; Ole Skøtt; Boye L. Jensen

To examine the role of the calcium/calmodulin-dependent phosphatase calcineurin in regulation of renin release, we assayed exocytosis using whole-cell patch clamp of single juxtaglomerular cells in culture. The calcineurin inhibitor, cyclosporine A (CsA), significantly increased juxtaglomerular cell membrane capacitance, an index of cell surface area and an established measure of exocytosis in single-cell assays. This effect was mimicked by intracellular delivery of a calcineurin inhibitory peptide, the calcium chelator ethylene glycol tetraacetic acid (EGTA), or the calmodulin inhibitor W-13. Simultaneous exposure to EGTA and CsA had no additive effect. The protein kinase A (PKA) blocker RpcAMPs had no effect on the CsA-induced increase in membrane capacitance. Intra- and extracellular application of tacrolimus did not alter membrane capacitance. A calmodulin antagonist (calmidazolium) and CsA, but not tacrolimus, significantly stimulated renin release from cultured juxtaglomerular cells. Juxtaglomerular cells expressed the calcineurin isoforms A-beta and A-gamma but not A-alpha. Plasma renin concentrations (PRCs) were not different in wild-type, calcineurin A-alpha, or A-beta knockout mice but increased after CsA treatment of the A-alpha knockout, while renin mRNA was suppressed. We conclude that calcineurin and calcium/calmodulin suppress exocytosis of renin from juxtaglomerular cells independent of PKA.

Collaboration


Dive into the Kirsten Madsen's collaboration.

Top Co-Authors

Avatar

Boye L. Jensen

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar

Niels Marcussen

Odense University Hospital

View shared research outputs
Top Co-Authors

Avatar

Ole Skøtt

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anne Robdrup Tinning

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar

Gitte Kjærsgaard

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar

Jane Stubbe

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ulla G. Friis

University of Southern Denmark

View shared research outputs
Researchain Logo
Decentralizing Knowledge