You are on page 1of 9

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/8574658

Inhibition of erythrocyte phosphatidylserine exposure by urea and Cl(-)

Article  in  American journal of physiology. Renal physiology · July 2004


DOI: 10.1152/ajprenal.00263.2003 · Source: PubMed

CITATIONS READS

69 454

10 authors, including:

Svetlana Mysina valérie Tanneur


University of Chester Centre Hospitalier Universitaire de Nice
27 PUBLICATIONS   2,699 CITATIONS    33 PUBLICATIONS   1,907 CITATIONS   

SEE PROFILE SEE PROFILE

Andreas F Mack Thomas Wieder


University of Tuebingen University of Tuebingen
155 PUBLICATIONS   5,291 CITATIONS    200 PUBLICATIONS   11,225 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Erythrocyte ‘Apoptosis’ View project

Pathways of cancer cell apoptosis View project

All content following this page was uploaded by Christophe Duranton on 11 May 2017.

The user has requested enhancement of the downloaded file.


Am J Physiol Renal Physiol 286: F1046 –F1053, 2004;
10.1152/ajprenal.00263.2003.

Inhibition of erythrocyte phosphatidylserine exposure by urea and Cl⫺


Karl S. Lang,1 Swetlana Myssina,1 Philipp A. Lang,1 Valerie Tanneur,1 Daniela S. Kempe,1
Andreas F. Mack,2 Stephan M. Huber,1 Thomas Wieder,1 Florian Lang,1 and Christophe Duranton1
Departments of 1Physiology and 2Anatomy, University of Tübingen, D-72076 Tübingen, Germany
Submitted 19 December 2003; accepted in final form 12 January 2004

Lang, Karl S., Swetlana Myssina, Philipp A. Lang, Valerie which opens Ca2⫹-permeable cation conductance (33). More-
Tanneur, Daniela S. Kempe, Andreas F. Mack, Stephan M. over, osmotic shock stimulates a sphingomyelinase (Smase)
Huber, Thomas Wieder, Florian Lang, and Christophe Duranton. with subsequent formation of ceramide (36), which potentiates
Inhibition of erythrocyte phosphatidylserine exposure by urea and the effect of cytosolic Ca2⫹ activity on erythrocyte scramblase
Cl⫺. Am J Physiol Renal Physiol 286: F1046 –F1053, 2004. 10.1152/
(33, 36).
ajprenal.00263.2003.—Osmotic shock by addition of sucrose to the
medium stimulates erythrocyte sphingomyelinase with subsequent Because osmotic shock may trigger Ca2⫹ entry and cer-
amide formation, the question arises whether those events

Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on May 11, 2017


ceramide formation and triggers Ca2⫹ entry through stimulation of
cation channels. Both ceramide and Ca2⫹ activate an erythrocyte occur during passage of erythrocytes through kidney medulla
scramblase, leading to breakdown of phosphatidylserine asymmetry, a with the extraordinary NaCl and urea required to concentrate
typical feature of apoptosis. Because erythrocytes are regularly ex- the urine (49). The present study has been performed to test
posed to osmotic shock during passage of kidney medulla, the present whether hypertonic NaCl and urea similarly induce annexin
study explored the influence of NaCl and urea on erythrocyte phos- binding as hypertonic sucrose and to elucidate the related
phatidylserine exposure as determined by annexin binding. The per- mechanisms.
centage of annexin-binding erythrocytes increased from ⬍5 to 80 ⫾
4% (n ⫽ 4) upon addition of 650 mM sucrose, an effect paralleled by METHODS
activation of the cation channel and stimulation of ceramide forma-
tion. The number of annexin-binding erythrocytes increased only to Solutions. Erythrocytes were drawn from healthy volunteer donors
18% after addition of 325 mM NaCl and was not increased by who gave informed consent, and the study was approved by the
addition of 650 mM urea. According to whole cell patch-clamp Ethical Committee of the Medical Faculty. Experiments were per-
experiments, the cation conductance was activated by replacement of formed at 37°C in a standard Ringer solution containing (in mM): 125
extracellular Cl⫺ with gluconate at isotonic conditions or by addition NaCl, 5 KCl, 1 MgSO4, 32 HEPES/NaOH, 5 glucose, and 1 CaCl2
of hypertonic sucrose or urea. Although stimulating the cation con- (pH ⫽ 7.4). Where indicated, osmolarity was increased by adding
ductance, urea abrogated the annexin binding and concomitant in- sucrose, NaCl, or urea on top of isotonic Ringer. In some experiments,
crease of ceramide levels induced by osmotic cell shrinkage. In vitro extracellular Cl⫺ was removed by replacing NaCl, KCl, and CaCl2
sphingomyelinase assays demonstrated a direct inhibitory effect of from the standard Ringer solution with equimolar amounts of sodium-
urea on sphingomyelinase activity. Urea did not significantly interfere D-gluconate (125 mM), potassium-D-gluconate (5 mM), and calcium-
with annexin binding after addition of ceramide. In conclusion, both (D-gluconate)2 (1 mM) or by NaNO3 (125 mM), KNO3 (5 mM), and
Cl⫺ and urea blunt erythrocyte phosphatidylserine exposure after Ca(NO3)2 (1 mM).
osmotic shock. Whereas Cl⫺ is effective through inhibition of the Smase from Staphylococcus aureus was purchased from Biomol
cation conductance, urea exerts its effect through inhibition of sphin- and used at a final concentration of 0.005 U/ml by serial dilution from
gomyelinase, thus blunting formation of ceramide. a 100 U/ml stock solution. Externally added Smase has been shown to
be capable of degrading cell membrane sphingomyelin with the
cation channels; sphingomyelinase; ceramide; apoptosis; cell volume; formation of ceramide (27, 43, 46, 51). D-Erythro-N-hexanoylsphin-
osmolarity; phosphatidylserine gosine (C6-ceramide) was purchased from Biomol (Hamburg, Ger-
many). D-Erythro-N-hexanoylsphingosine was dissolved in DMSO to
give a 50 mM stock solution and further diluted to a final concentra-
ERYTHROCYTES LACK MITOCHONDRIA and nuclei, intracellular or-
tion of 50 ␮M in standard Ringer solution containing 0.1% BSA. The
ganelles involved in the induction of apoptosis (22, 24). Nev- maximum concentration of DMSO was in all cases 0.1%, a concen-
ertheless, treatment of erythrocytes with the Ca2⫹ ionophore tration that did not induce annexin binding (data not shown).
ionomycin has recently been shown to induce erythrocyte A monoclonal anti-ceramide antibody (clone MID 15B4; isotype
shrinkage, membrane blebbing, and breakdown of cell mem- IgM) was purchased from Alexis (Grünberg, Germany).
brane phosphatidylserine asymmetry, all typical features of FACS analysis. FACS analysis was performed essentially as de-
apoptosis in other cell types (5, 10, 15, 33). Cells exposing scribed earlier (2, 31). After incubation, cells were washed in annexin-
phosphatidylserine at their surface are recognized by phospha- binding buffer containing 125 mM NaCl, 10 mM HEPES/NaOH
tidylserine receptor-carrying macrophages (20), which bind, (pH ⫽ 7.4), and 5 mM CaCl2. Erythrocytes were stained with
annexin-FLUOS (Böhringer Mannheim, Mannheim, Germany) at a
phagocytose, and thus clear those cells from circulating blood.
1:100 dilution. After 15 min, samples were diluted 1:5 and measured
The breakdown of phosphatidylserine asymmetry results by flow cytometric analysis on a FACS-Calibur (Becton-Dickinson,
from the stimulation of a scramblase (63), which is activated Heidelberg, Germany). Cells were analyzed by forward and side
by an increase of cytosolic Ca2⫹ activity (16, 58, 59). Cyto- scatter, and annexin-fluorescence intensity was measured in FL-1.
solic Ca2⫹ activity is increased during exposure of erythrocytes Control experiments revealed that urea (600 mM) did not interfere
to osmotic shock by an increase of extracellular osmolarity, with the annexin-binding assay of hypertonically stressed erythrocytes

Address for reprint requests and other correspondence: F. Lang, Physiolo- The costs of publication of this article were defrayed in part by the payment
gisches Institut, der Universität Tübingen, Gmelinstr. 5, D-72076 Tübingen of page charges. The article must therefore be hereby marked “advertisement”
(E-mail: florian.lang@uni-tuebingen.de). in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

F1046 0363-6127/04 $5.00 Copyright © 2004 the American Physiological Society http://www.ajprenal.org
INHIBITION OF ERYTHROCYTE “APOPTOSIS” BY UREA AND CL⫺ F1047
(Ringer ⫹ 650 mM sucrose for 8 h) when added 5 min before the end presence or absence of 600 mM urea). Samples were sonicated for 5
of incubation. In those experiments, the percentage of annexin- min, and 0.2 U/ml Smase from Staphylococcus aureus (Biomol) or
binding cells was 63 ⫾ 6% (n ⫽ 6) in the absence and 62 ⫾ 5% Streptomyces species (Sigma, Taufkirchen, Germany) were added.
(n ⫽ 6) in the presence of urea. Reaction mixtures were incubated for different times at 37°C. After 5,
For determination of ceramide formation, cells were stained for 1 h 10, and 30 min, 20 ␮l of the samples were removed, and the reactions
at 4°C with 1 ␮g/ml anti-ceramide antibody in PBS containing 1% were stopped by addition of 133 ␮l chloroform and 67 ␮l methanol.
FCS at a dilution of 1:5, as described recently (36). After three washes Phase separation was completed by addition of 20 ␮l water. Smase-
with PBS/1% FCS, cells were stained with polyclonal FITC-conju- catalyzed release of [3H]phosphocholine was quantitated by counting
gated goat anti-mouse Ig-specific antibody (Pharmingen, Hamburg, 20 ␮l of the upper, aqueous phase. Blank reactions contained no
Germany) in PBS/1% FCS at a dilution of 1:50 for 30 min. Unbound Smase, and values were subtracted from the sample values.
secondary antibody was removed by washing the cells two times Patch clamp. Patch-clamp experiments have been performed as
with PBS/1% FCS, and samples were analyzed by flow cytometric described earlier (26, 36). Erythrocytes were recorded at 21°C. A
analysis on a FACS-Calibur. FITC-fluorescence intensity was mea- continuous superfusion was applied through a flow system inserted in
sured in FL-1. the dish. The bath was grounded via an NaCl bridge filled with bath
Immunofluorescence. Control or ischemic (30 min) kidneys were NaCl solution (see below). Borosilicate glass pipettes (8 –14 M⍀ tip
quickly frozen in liquid nitrogen and transferred to a cryostat. Exper- resistance; GC 150 TF-10; Clark Medical Instruments, Pangbourne,
iments were performed according to the German Animal Protection UK) manufactured by a microprocessor-driven DMZ puller (Zeitz,

Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on May 11, 2017


Law and were approved by the local authorities. Sections were taken Augsburg, Germany) were used in combination with an MS314
at 20 ␮m and stained unfixed with annexin-V-FLUOS (Roche) diluted electrical micromanipulator (MW; Märzhäuser, Wetzlar, Germany).
1:100 in buffer containing (in mM) 140 NaCl, 10 HEPES/NaOH, and The currents were recorded in voltage-clamp mode in fast whole cell
5 CaCl2 (pH 7.4). After being rinsed two times with the same buffer, configuration by an EPC-9 amplifier (Heka, Lambrecht, Germany)
sections were covered with a coverslip and analyzed immediately. using Pulse software (Heka) and an ITC-16 Interface (Instrutech, Port
Sections were examined on a Zeiss LSM510 confocal microscope Washington, NY). The whole cell currents were evoked by a pulse
using an Argon laser with excitation at 488 nm, with appropriate protocol, clamping the voltage in 11 successive 400-ms square pulses
emission filters, and a ⫻40 oil immersion lens (numerical aperture from the ⫺10 mV holding potential to potentials between ⫺100 and
1.3). Images or small z-stacks of images from large blood vessels in ⫹100 mV.
the outer medulla were scanned, and the number of annexin-V- The potentials were corrected for liquid junction potentials (3). The
FLUOS-positive cells was counted. The confocal microscopy soft- original whole cell current traces are depicted after 500 Hz low-pass
ware supplied by the manufacturer was used to measure the cross- filtering, and currents of the individual voltage square pulses are
sectioned area of the blood vessels. Together with the optical section superimposed. The applied voltages refer to the cytoplasmic face of
thickness or z-dimension of the stack (ranging from 3 to 6 ␮m), we the cell membrane with respect to the extracellular space. The inward
calculated the number of stained cells per volume. currents, defined as flow of positive charge from the extracellular to
Measurement of intracellular ATP concentrations. The intracellu- the cytoplasmic membrane face, are negative currents and depicted as
lar ATP concentration of human erythrocytes was quantified by a downward deflections of the original current traces.
luciferin-luciferase assay kit (Roche Diagnostics, Mannheim, Ger- Whole cell currents were recorded in standard isotonic bath solu-
many) using a luminometer (Berthold Biolumat LB9500, Bad Wild- tion containing (in mM) 115 NaCl, 10 HEPES, 5 CaCl2, and 10
bad, Germany). Briefly, fresh erythrocytes were washed (3 ⫻ 5 min) MgCl2, titrated with NaOH to pH 7.4, in combination with a sodium-
in PBS Ca2⫹-free medium and centrifuged, and 100 ␮l of blood pellet D-gluconate or potassium-D-gluconate pipette solution containing (in
(hematocrit 100%) were incubated for 8 h at 37°C with standard mM) 115 sodium-D-gluconate or potassium-D-gluconate, 10 NaCl, 1
Ringer solution or Ringer supplemented with 650 mM urea or 650 EGTA, 1 MgCl2, 1 Mg-ATP, and 5 HEPES, titrated to pH 7.35 with
mM sucrose (final hematocrit ⬃5–7%). After incubation, cells were NaOH. A residual Cl⫺ concentration of 10 mM was used in these
lysed in distillated water, and proteins were precipitated by HClO4 pipette solutions to avoid shifts in offset potential.
(5%). After centrifugation, an aliquot of the supernatant (400 ␮l) was Where indicated, NaCl bath solution was replaced by sodium-D-
adjusted to pH 7.7 by addition of saturated KHCO3 solution. All of the gluconate solution containing (in mM): 140 sodium gluconate, 5
manipulations were performed at 4°C to avoid ATP degradation. HEPES, 1 CaCl2, and 1 MgCl2. The removal of Cl⫺ eliminates the
After dilution of the supernatant, the ATP concentration of the outward currents generated by Cl⫺ influx and upregulates the cation
aliquots was determined by luciferin/luciferase reaction according to conductance (18). For some experiments, the osmolarity of the so-
the manufacturer’s protocol. The given values are representative of dium-D-gluconate bath solution was increased by addition of 250 mM
the average ATP concentration measured for 100 ␮l pelleted blood sucrose or 250 mM urea.
(hematocrit 100%). Statistics. Data are expressed as arithmetic means ⫾ SE. Statistical
In vitro Smase assay. In vitro measurements of Smase activity were analysis was made by paired or unpaired t-test, where appropriate.
performed essentially as described earlier (36) using total lipid ex-
tracts from erythrocytes supplemented with [choline-methyl- RESULTS
3
H]sphingomyelin (with a specific radioactivity of 2.96 TBq/mmol;
purchased from American Radiolabeled Chemicals, St. Louis, MO) as Effects of hypertonic sucrose, NaCl, and urea on erythrocyte
substrate. Erythrocytes (2 ⫻ 109) were washed two times with 1 ml annexin binding. In isotonic extracellular fluid, only a small
standard Ringer solution. Next, total lipids were extracted by resus- fraction of human erythrocytes (4 ⫾ 1%, n ⫽ 12) showed
pending the cell pellets in 500 ␮l methanol, 250 ␮l chloroform, and detectable annexin binding, reflecting the virtual absence of
200 ␮l water. Samples were stirred for 10 min on a vortex mixer and phosphatidylserine at the extracellular face of the erythrocyte
centrifuged at 13,000 g for 2 min. Phase separation was accomplished cell membrane (Fig. 1A). Increase of osmolarity by addition of
by the addition of 250 ␮l chloroform and 250 ␮l water. The suspen-
sion and centrifugation steps were repeated. [choline-methyl-
sucrose, however, led to a sharp increase in annexin binding,
3
H]sphingomyelin (55.5 kBq) was added to 150 ␮l of the chloroform reflecting breakdown of the phosphatidylserine asymmetry
phase and stirred for 1 min. The radioactive substrate solution (10 ␮l) (Fig. 1A); after addition of 650 mM sucrose, 80 ⫾ 4% (n ⫽ 4)
was dried under nitrogen and used for the in vitro Smase assay. The of the human erythrocytes bound annexin within 8 h (Fig. 1, A
radioactive substrate was resuspended in 100 ␮l of assay buffer (100 and B). The same increase in osmolarity by addition of 325
mM Tris·HCl, pH 7.4, 6 mM MgCl2, and 0.1% Triton X-100 in the mM NaCl (650 mosmol/l) induced annexin binding only in
AJP-Renal Physiol • VOL 286 • JUNE 2004 • www.ajprenal.org
F1048 INHIBITION OF ERYTHROCYTE “APOPTOSIS” BY UREA AND CL⫺

tration compared with the control condition (isosmotic Ringer,


n ⫽ 15, Fig. 1C).
Inhibitory effect of urea on annexin binding in erythrocytes
exposed to osmotic shock. The inability of hypertonic urea to
induce annexin binding prompted us to explore whether urea
could inhibit the annexin binding of erythrocytes exposed to
hypertonic sucrose concentrations. In the absence of urea, the
addition of sucrose (Ringer ⫹ 650 mM) led within 8 h to
annexin binding in 73 ⫾ 5% of the cells (n ⫽ 4; Fig. 2A). The
addition of urea (600 mM) blunted the sucrose-induced an-
nexin binding despite a further increase of osmolarity (Fig.
2A). In contrast, an increase to the same osmolarity by addition
of sucrose (1,250 mM) induced annexin binding in 80 ⫾ 3%
(n ⫽ 6) of the cells. Figure 2B shows the dose-response curve
for urea-dependent inhibition of erythrocyte annexin binding at

Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on May 11, 2017


a constant concentration of sucrose (650 mM on top of isotonic
Ringer) and increasing concentrations of urea (added on top of
isotonic Ringer and sucrose). The additional presence of 600
mM urea decreased the percentage of annexin-binding cells to
20 ⫾ 5% (n ⫽ 4, Fig. 2, A and B) despite the presence of 650
mM sucrose on top of isotonic Ringer. Although the addition
of up to 300 mM sucrose on top of isotonic Ringer does not
significantly increase the number of annexin-binding cells (Fig.
1B), the addition of 250 mM sucrose and replacement of NaCl
with sodium gluconate significantly enhanced the number of
annexin-binding cells, an effect partially reversed by further
addition of urea (600 mM, Fig. 2C).
Because urea, unlike sucrose, rapidly enters cells (32), it
Fig. 1. Effect of hypertonic sucrose, NaCl, and urea on erythrocyte annexin does not create an osmotic gradient across the cell membrane.
binding and intracellular ATP. A: histograms showing the percentage of We thus explored whether phloretin, a nonspecific inhibitor of
annexin-binding erythrocytes after incubation for 8 h in isotonic Ringer urea transport (41), modifies the effect of urea on annexin
solution (top left), Ringer solution ⫹ 650 mM sucrose (top right), Ringer
solution ⫹ 325 mM NaCl (bottom left), or Ringer solution ⫹ 650 mM urea binding. Phloretin (700 ␮M) led to a slight increase of annexin
(bottom right). Nos. refer to the calculated percentage of one single experi- binding in isotonic Ringer (9 ⫾ 2%, n ⫽ 9) but did not
ment. B: percentage of annexin-binding erythrocytes after 8 h of treatment with significantly interfere with the inhibitory effect of urea. Urea
urea, NaCl, and sucrose as a function of osmolarity (arithmetic means ⫾ SE, (600 mM) reduced the annexin binding induced by hyperos-
n ⫽ 4). The cells were suspended in Ringer NaCl solution (300 mosmol/l) with
additional increasing concentrations of sucrose (from 300 to 650 mM), NaCl
motic shock (Ringer plus 650 mM sucrose for 8 h) by 44 ⫾ 8%
(from 150 to 325 mM), or urea (from 300 to 650 mM). C: histograms showing (n ⫽ 9) and 52 ⫾ 8% (n ⫽ 9) in the presence and absence of
the concentrations of intracellular ATP after incubation for 8 h in isotonic phloretin, respectively.
Ringer solution, Ringer solution ⫹ 650 mM sucrose, or Ringer solution ⫹ 650 In the absence of extracellular Ca2⫹, the effect of exposure
mM urea (arithmetic means ⫾ SE, n ⫽ 15). Data are expressed as mM to hypertonic shock (Ringer ⫹ 650 mM sucrose) on annexin
intracellular ATP/l blood (hematocrit ⬃100%, n ⫽ 10).
binding was significantly blunted by 17 ⫾ 6% (n ⫽ 5), but the
inhibitory effect of urea was preserved. The percentage of
inhibition was 57 ⫾ 5% (n ⫽ 5) in the absence of Ca2⫹
18 ⫾ 3% (n ⫽ 4) of the cells (Fig. 1, A and B). In sharp compared with 58 ⫾ 5% (n ⫽ 5) in the presence of Ca2⫹.
contrast, addition of 650 mM urea did not significantly enhance Activation of cation conductance by Cl⫺ removal and hy-
the percentage of annexin-binding cells (4 ⫾ 1%, n ⫽ 4; Fig. perosmolarity. Patch-clamp experiments have been performed
1, A and B). As shown in Fig. 1B, a correlation is observed to elucidate the role of osmolarity, Cl⫺, and urea in the
between the number of annexin-binding cells and the hyper- regulation of the nonselective cation conductance. As shown in
osmolarity by increasing concentrations of sucrose (varying Fig. 3A, the cation conductance was activated by replacement
from 300 to 650 mM) or NaCl (varying from 150 to 325 mM) of NaCl with sodium gluconate, even in isotonic extracellular
but not urea (varying from 300 to 650 mM). The annexin fluid. Upon removal of external Cl⫺, the conductance (G; as
binding after osmotic shock was not significantly blunted in the calculated between ⫹40 and ⫹100 mV) was 223 ⫾ 41 pS
presence of the Na⫹-K⫹-2Cl⫺ cotransport inhibitor bumet- (n ⫽ 17) and 251 ⫾ 51 pS (n ⫽ 8) when recorded with sodium
anide. In this series of experiments, after 8 h of osmotic shock gluconate and potassium gluconate pipette solution, respec-
(Ringer ⫹ 650 mM sucrose), the percentage of annexin- tively.
binding cells was 45 ⫾ 2 (n ⫽ 4) in the absence and 46 ⫾ 2 Addition of either sucrose (250 mM) or urea (250 mM) on
(n ⫽ 4) in the presence of bumetanide (100 ␮M). top of isotonic sodium gluconate further stimulated the chan-
ATP measurements using luciferin-luciferase assay revealed nel. Figure 3B depicts the corresponding current-voltage rela-
that an 8-h incubation in Ringer supplemented with 650 mM tionships recorded with isosmotic sodium gluconate (n ⫽ 16),
urea or 650 mM sucrose induced a moderate decrease (8 and sodium gluconate ⫹ 250 mM sucrose (n ⫽ 4), or sodium
32%, respectively, n ⫽ 15) of the intracellular ATP concen- gluconate ⫹ 250 mM urea (n ⫽ 4). Figure 3C shows the mean
AJP-Renal Physiol • VOL 286 • JUNE 2004 • www.ajprenal.org
INHIBITION OF ERYTHROCYTE “APOPTOSIS” BY UREA AND CL⫺ F1049

Fig. 2. Inhibition of sucrose-induced annexin binding by


urea. A: histograms of annexin binding in a representa-
tive experiment of erythrocytes incubated in isotonic
Ringer solution (left), in Ringer solution ⫹ 650 mM
sucrose (suc; middle), and in Ringer solution ⫹ 650 mM
sucrose ⫹ 600 mM urea (right) for 8 h. B: percentage of
annexin-binding erythrocytes after 8 h of incubation
with a Ringer solution ⫹ 650 mM sucrose as a function
of the concentration of urea added on top of 650 mM
sucrose (arithmetic means ⫾ SE, n ⫽ 4). C: percentage
of annexin-binding erythrocytes after 8 h of incubation
with Cl⫺-free Ringer solution (NaCl replaced by so-

Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on May 11, 2017


dium gluconate) ⫹ 250 mM sucrose as a function of the
concentration of urea added on top of 250 mM sucrose
(arithmetic means ⫾ SE, n ⫽ 4).

slope conductance (calculated between ⫹40 and ⫹100 mV) Because removal of extracellular Cl⫺ activates the cation
recorded with isosmotic sodium gluconate bath solution (G ⫽ conductance even without an increase of osmolarity, the effect
0.31 ⫾ 0.06 nS, n ⫽ 16) and after increasing the osmolarity of of Cl⫺ removal on annexin binding was tested. As shown in
the sodium gluconate bath solution by addition of 250 mM Fig. 3D, replacement of isotonic Cl⫺ by isotonic gluconate
sucrose (G ⫽ 0.52 ⫾ 0.04 nS, n ⫽ 4) or 250 mM urea (G ⫽
0.47 ⫾ 0.06 nS, n ⫽ 4). Thus, similar to hypertonic sucrose,
hypertonic urea activates the cation conductance.

Fig. 3. Activation of cation channels by Cl⫺ removal and hyperosmolarity. A:


original whole cell current traces (sodium gluconate pipette solution) recorded
with isotonic bath solution (NaCl), after removal of Cl⫺ (sodium gluconate),
addition of 250 mM sucrose (sodium gluconate ⫹ sucrose), replacement of
sucrose by 250 mM urea (sodium gluconate ⫹ urea), and washout of urea by
isotonic bath solution (NaCl). B: current (I)-voltage (V) relationships recorded
as in A with isotonic (‚) and hypertonic sodium gluconate bath solution
(sodium gluconate ⫹ 250 mM sucrose; {) and after isosmotic replacement of Fig. 4. Inhibition of sphingomyelinase (Smase)-induced annexin binding by
sucrose by urea (sodium gluconate ⫹ 250 mM urea, }). Data are means ⫾ SE urea. A: representative histograms of erythrocytes incubated for 8 h in isotonic
(n ⫽ 4). C: comparison of the mean conductance ⫾ SE (n ⫽ 4) calculated from Ringer solution with purified Smase (0.005 U/ml, top left), Smase (0.005
the outward current (⫹40 to ⫹100 mV) in the presence of isotonic sodium U/ml) ⫹ urea (600 mM, top right), ceramide (50 ␮M, bottom left), or ceramide
gluconate bath solution or under hypertonic conditions after addition of 250 (50 ␮M) ⫹ urea (600 mM, bottom right). B: arithmetic means ⫾ SE (n ⫽ 4)
mM sucrose or 250 mM urea to the sodium gluconate bath solution. D: annexin of the percentage of annexin-binding cells stimulated with Smase (0.005 U/ml)
binding of erythrocytes after a 20-h treatment in isotonic extracellular fluid or ceramide (50 ␮M) for 8 h in the presence of different urea concentrations.
without and with isosmotic replacement of NaCl by sodium gluconate (arith- Control refers to cells stimulated with Smase (0.005 U/ml) or ceramide (50
metic means ⫾ SE, n ⫽ 4). ␮M) in the absence of urea.

AJP-Renal Physiol • VOL 286 • JUNE 2004 • www.ajprenal.org


F1050 INHIBITION OF ERYTHROCYTE “APOPTOSIS” BY UREA AND CL⫺

within 20 h significantly increased the number of annexin-


binding cells. Similarly, replacement of extracellular Cl⫺
by isotonic NO⫺ 3 significantly increased the number of
annexin-binding cells from 5 ⫾ 1% (n ⫽ 10) to 11 ⫾ 2%
(n ⫽ 10).
Inhibitory effect of urea on Smase-induced annexin binding.
Because urea inhibited annexin binding despite the activation
of the nonselective cation conductance, experiments were per-
formed to test for a putative effect of urea on Smase-mediated
annexin binding. As shown in Fig. 4A, exposure of erythro-
cytes to purified Smase (0.005 U/ml) led within 8 h to annexin
binding in 68 ⫾ 2% (n ⫽ 4) of the cells. A similar increase of
the percentage of annexin-binding cells (62 ⫾ 11%, n ⫽ 4) was
observed after C6-ceramide exposure (50 ␮M). Figure 4B
shows the concentration-dependent effect of urea on Smase-

Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on May 11, 2017


induced annexin binding. In these experiments, addition of 600
mM urea virtually abolished (3 ⫾ 2%, n ⫽ 4) the breakdown
of phosphatidylserine asymmetry after addition of Smase. In
contrast, ceramide-induced annexin binding was further en-
hanced in the presence of urea, approaching 95 ⫾ 3% (n ⫽ 4)
of the cells for 600 mM urea (Fig. 4B).
Inhibition of ceramide formation and Smase activity by urea.
The observation that urea inhibits annexin binding after addi-
tion of Smase and hypertonicity, but not after addition of
ceramide, suggested that urea directly influences Smase. Fur-
ther experiments have been performed to test for an effect of
urea on Smase activity. As shown in Fig. 5, osmotic shock
induced by addition of sucrose (650 mM sucrose) stimulated

Fig. 6. Inhibition of in vitro Smase activity by urea. A: time-dependent release


of [3H]phosphocholine from [choline-methyl-3H]sphingomyelin by Smase was
measured in control assay buffer as described in METHODS. Arithmetic means ⫾
SE (n ⫽ 3) of liberated [3H]phosphocholine are given in dpm. B: arithmetic
means ⫾ SE (n ⫽ 3) of liberated [3H]phosphocholine/min after incubation in
assay buffer in the absence (control) or in the presence of 600 mM urea (urea).
Smase either from Staphylococcus aureus or Streptomyces species was used as
an enzyme source. *Significant difference from control assay buffer (2-tailed
t-test, P ⱕ 0.05).

ceramide formation in erythrocytes (the levels of ceramide


formation start to increase after 30 min and reach a plateau
after 4 h; data not shown). After 4 h, ceramide levels reached
181 ⫾ 30% of control (n ⫽ 4), an effect completely reversed
to 106 ⫾ 15% of control (n ⫽ 4) in the presence of urea (650
mM sucrose ⫹ 600 mM urea). As a positive control for
ceramide detection, erythrocytes were treated with purified
Fig. 5. Inhibition of ceramide formation by urea. A: representative histograms
Smase (1 U/ml during 5 min). As expected, this maneuver led
of ceramide detection in control cells (isotonic Ringer, black, all histograms) to increased ceramide formation, approaching 303 ⫾ 48% (n ⫽
and cells exposed for 4 h to hypertonic sucrose solution (Ringer ⫹ 650 mM 4) of control (Fig. 5B).
sucrose) in the absence (gray, left) or in the presence (gray, middle) of 600 mM In vitro assays were performed in addition to test for the
urea. Cells exposed for 5 min to purified Smase in isotonic Ringer (1 U/ml) influence of urea on Smase activity. For this, total erythrocyte
served as a positive control for ceramide production (gray, right). B: arithmetic
means ⫾ SE (n ⫽ 4) of the mean ceramide-related fluorescence after 4 h of lipid extract supplemented with [choline-methyl-3H]sphingo-
incubation with an isotonic Ringer NaCl solution (control), after 4 h in myelin was used as substrate. As shown in Fig. 6A, production
hypertonic sucrose solution (suc; Ringer ⫹ 650 mM sucrose), after 4 h in of [3H]phosphocholine increased within 10 min of incubation
hypertonic sucrose solution in the presence of urea (suc ⫹ urea, Ringer ⫹ 650 with Smase and could thus serve as a direct measure of Smase
mM sucrose ⫹ 600 mM urea), and after 5 min in the presence of 1 U/ml
purified Smase in isotonic Ringer (Smase) as a positive control. *Significant
activity. Urea significantly reduced the activity of Smase from
difference from control cells and #significant difference from cells in hyper- Staphylococcus aureus and Streptomyces species by 44 ⫾ 4%
tonic sucrose solution in the absence of urea (2-tailed t-test, P ⱕ 0.05). (n ⫽ 3) and 35 ⫾ 2% (n ⫽ 3), respectively (Fig. 6B).
AJP-Renal Physiol • VOL 286 • JUNE 2004 • www.ajprenal.org
INHIBITION OF ERYTHROCYTE “APOPTOSIS” BY UREA AND CL⫺ F1051
Induction of erythrocyte phosphatidylserine exposure in which may at least in part result from enhanced ATP hydro-
ischemic mouse kidney. To test for a possible role in acute renal lysis (60). A decrease of ATP concentration may interfere with
failure, erythrocyte phosphatidylserine exposure and vessel Ca2⫹ extrusion by the Ca2⫹-ATPase and thus increase cyto-
wall adherence were estimated in control and ischemic (30 solic Ca2⫹ concentration.
min) mouse kidney by fluorescence microscopy. As shown in Beyond the activation of nonspecific cation conductance,
Fig. 7, A and B, ischemia induced a moderate but significant hyperosmotic cell shrinkage activates in erythrocytes an endo-
(P ⱕ 0.05) increase in the percentage of phosphatidylserine- geneous Smase, leading within 30 min to the formation of
exposing erythrocytes (from 100 ⫾ 23 to 231 ⫾ 45%; n ⫽ ceramide and with a slight delay to stimulation of annexin
6 – 8; Fig. 7C). Specifically, phosphatidylserine-exposing binding (36). The formation of ceramide is paralleled by the
erythrocytes were found in the large veins at the cortex- decrease of sphingomyelin (36). External application of cer-
medulla interface. In particular, a fraction of phosphatidyl- amide triggers annexin binding in erythrocytes (36) and nucle-
serine-exposing erythrocytes was adjacent to the vessel wall, ated cells (17, 38). Ceramide sensitizes erythrocytes for the
suggesting cytoadherence to the endothelium. action of Ca2⫹ (36). Along those lines, the increase of annexin
binding after osmotic shock was blunted in the presence of the
DISCUSSION
putative Smase inhibitor dichloroisocoumarin (36). In nucle-

Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on May 11, 2017


The present study confirms the previous observations that ated cells (27, 43) and erythrocytes, addition of exogenous
hyperosmotic shock stimulates breakdown of the phosphati- Smase similarly stimulates ceramide formation and subsequent
dylserine asymmetry in erythrocytes (33, 35). In vivo, nucle- increase of annexin binding. Involvement of ceramide in sig-
ated cells (20) and erythrocytes (6, 19) exposing phosphatidyl- naling of apoptosis has been shown for nucleated cells, such as
serine will be eliminated by macrophages. As shown previ- CD95-induced lymphocyte death (23, 25) and treatment of
ously, hyperosmotic shock and oxidative stress open a cells with various chemotherapeutic agents, e.g., hexade-
nonselective cation conductance allowing the passage of Ca2⫹ cylphosphocholine (57) or daunorubicin (9). The ability of
(18, 26). Volume-regulatory cation conductances are not only C6-ceramide to induce erythrocyte phosphatidylserine expo-
expressed in erythrocytes but also in a wide variety of nucle- sure is surprising, since erythrocytes lack mitochondria, crucial
ated cells (11, 12, 21, 30, 32, 54 –56). The cation conductance elements in the signaling cascade triggered by C6-ceramide in
in erythrocytes has been characterized previously (18, 26). In nucleated cells. C6-ceramide must utilize a pathway distinct
those cells, the cation conductance probably does not serve cell from that described in nucleated cells. Thus the capacity of
volume regulation. In contrast, Ca2⫹ entry through the cation C6-ceramide to trigger phosphatidylserine exposure in eryth-
conductance (33) activates the Gardos channel (35), leading to rocytes discloses the presence of an alternative pathway that
hyperpolarization, loss of cellular KCl, and cell shrinkage. may exist in nucleated cells as well. C6-ceramide triggers
Increase of extracellular K⫹ rather blunts cell shrinkage and erythrocyte phosphatidylserine exposure even though it does
erythrocyte annexin binding after an increase of cytosolic not enhance Ca2⫹ uptake (36).
Ca2⫹ (34). Excessive osmolarity, as it prevails in renal medulla, may
Increase of cytosolic Ca2⫹ in the erythrocytes may result not trigger apoptosis of nucleated cells (7, 8, 32, 35, 40, 45, 47,
only from enhanced entry but also from impaired extrusion. 48). Moreover, it compromises the function of lymphocytes,
Osmotic shock leads to a decrease of ATP concentrations, which inhibits defense in kidney medulla (37). In cultured

Fig. 7. Annexin-binding erythrocytes in is-


chemic kidney. A and B: confocal laser scan
images from sections of control (A) and is-
chemic (30 min; B) mouse kidneys stained
with annexin-V-FLUOS to detect phosphati-
dylserine-exposing erythrocytes. Shown are
fluorescence images (left) and superimposed
fluorescence and transmission light images
(right) from veins at the cortex medulla bor-
der (as indicated by scheme on top right).
Arrowheads indicate phosphatidylserine-ex-
posing erythrocytes adhering at the vessel
wall. C: relative number of phosphatidyl-
serine-exposing erythrocytes in sections of
control (open bar) and ischemic (closed bar)
mouse kidneys. Data are means ⫾ SE of 6 – 8
z-stacks of vessel images from 2 kidneys for
each condition. OM, outer medulla; IM, in-
ner medulla.

AJP-Renal Physiol • VOL 286 • JUNE 2004 • www.ajprenal.org


F1052 INHIBITION OF ERYTHROCYTE “APOPTOSIS” BY UREA AND CL⫺

renal tubular epithelial cells, the apoptosis induced by NaCl 2. Andree HA, Reutelingsperger CP, Hauptmann R, Hemker HC, Her-
could be reversed by urea (53, 61, 62). On the other hand, urea mens WT, and Willems GM. Binding of vascular anticoagulant alpha
(VAC alpha) to planar phospholipid bilayers. J Biol Chem 265: 4923–
has been shown to sensitize cells to apoptosis (13) and lead to 4928, 1990.
cell cycle delay (45). In the present study, we clearly show that 3. Barry PH and Lynch JW. Liquid junction potentials and small cell
urea (600 mM) inhibits hyperosmotic shock-induced erythro- effects in patch-clamp analysis. J Membr Biol 121: 101–117, 1991.
cyte annexin binding. Notably, erythrocytes exposed to 300 4. Bensinger TA, Maisels MJ, Mahmood L, McCurdy PR, and Conrad
mM NaCl plus 600 mM urea for 8 h (which approximates the ME. Effect of intravenous urea in invert sugar on heme catabolism in
hyperosmotic condition in the medullary tip) only show mar- sickle-cell anemia. N Engl J Med 285: 995–997, 1971.
5. Berg CP, Engels IH, Rothbart A, Lauber K, Renz A, Schlosser SF,
ginal annexin binding of 11.4 ⫾ 4.5% (n ⫽ 8) compared with Schulze-Osthoff K, and Wesselborg S. Human mature red blood cells
erythrocytes exposed to isotonic Ringer solution (2.9 ⫾ 1.4%, express caspase-3 and caspase-8, but are devoid of mitochondrial regula-
n ⫽ 8). Thus the high Cl⫺ and urea concentrations prevailing tors of apoptosis. Cell Death Differ 8: 1197–1206, 2001.
in renal medulla prevent erythrocyte phosphatidylserine expo- 6. Boas FE, Forman L, and Beutler E. Phosphatidylserine exposure and
sure despite increased osmolarity. Under physiological condi- red cell viability in red cell aging and in hemolytic anemia. Proc Natl Acad
Sci USA 95: 3077–3081, 1998.
tions, the contact time of erythrocytes with renal medulla is too 7. Bortner CD and Cidlowski JA. A necessary role for cell shrinkage in
short to trigger significant phosphatidylserine exposure. How- apoptosis. Biochem Pharmacol 56: 1549 –1559, 1998.

Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on May 11, 2017


ever, the contact time may be considerably increased in acute 8. Bortner CD and Cidlowski JA. Caspase independent/dependent regula-
renal failure, which typically leads to trapping of erythrocytes tion of K(⫹), cell shrinkage, and mitochondrial membrane potential
in the transition from medulla to cortex (42). In the present during lymphocyte apoptosis. J Biol Chem 274: 21953–21962, 1999.
study, experimental renal ischemia, indeed, induced phospha- 9. Bose R, Verheij M, Haimovitz-Friedman A, Scotto K, Fuks Z, and
Kolesnick R. Ceramide synthase mediates daunorubicin-induced apopto-
tidylserine exposure of erythrocytes. Moreover, at least in sis: an alternative mechanism for generating death signals. Cell 82:
theory, urea could similarly interfere with ceramide formation 405– 414, 1995.
in renal medullary cells. 10. Bratosin D, Estaquier J, Petit F, Arnoult D, Quatannens B, Tissier JP,
The mechanisms by which urea interferes with apoptotic Slomianny C, Sartiaux C, Alonso C, Huart JJ, Montreuil J, and
signaling are poorly understood. Urea may activate ras (52) Ameisen JC. Programmed cell death in mature erythrocytes: a model for
investigating death effector pathways operating in the absence of mito-
and increase phosphatidylinositol 3-kinase activity (62). In chondria. Cell Death Differ 8: 1143–1156, 2001.
erythrocytes, urea has been shown to modify several transport 11. Cabado AG, Vieytes MR, and Botana LM. Effect of ion composition on
processes, including the Na⫹-K⫹-ATPase, KCl cotransport, the changes in membrane potential induced with several stimuli in rat mast
and Na⫹-K⫹-2Cl⫺ cotransport (28, 29, 39, 50). Interestingly, cells. J Cell Physiol 158: 309 –316, 1994.
hydroxyurea protects erythrocytes against oxidative damage 12. Chan HC, Goldstein J, and Nelson DJ. Alternate pathways for chloride
(1) and prevents sickling (4, 14, 44). In this respect, it may be conductance activation in normal and cystic fibrosis airway epithelial
cells. Am J Physiol Cell Physiol 262: C1273–C1283, 1992.
of interest that sickle cells are especially sensitive to phospha- 13. Colmont C, Michelet S, Guivarc’h D, and Rousselet G. Urea sensitizes
tidylserine exposure induced by osmotic shock, oxidative mIMCD3 cells to heat shock-induced apoptosis: protection by NaCl. Am J
stress, and energy depletion (33). The direct ability of urea to Physiol Cell Physiol 280: C614 –C620, 2001.
inhibit Smase and ceramide formation may well participate in 14. Culliford SJ, Ellory JC, Gibson JS, and Speake PF. Effects of urea and
the regulation of phosphatidylserine exposure not only in oxygen tension on K flux in sickle cells. Pflügers Arch 435: 740 –742,
1998.
erythrocytes but also in nucleated cells. 15. Daugas E, Cande C, and Kroemer G. Erythrocytes: death of a mummy.
In summary, similar to nucleated cells, erythrocytes loose Cell Death Differ 8: 1131–1133, 2001.
phosphatidylserine asymmetry, leading to annexin binding af- 16. Dekkers DW, Comfurius P, Bevers EM, and Zwaal RF. Comparison
ter exposure to hyperosmotic shock. The phosphatidylserine between Ca2⫹-induced scrambling of various fluorescently labelled lipid
exposure is mediated by both activation of cation conductance analogues in red blood cells. Biochem J 362: 741–747, 2002.
and by Smase-related ceramide formation. Cl⫺ directly inhibits 17. Dobrowsky RT and Hannun YA. Ceramide stimulates a cytosolic
protein phosphatase. J Biol Chem 267: 5048 –5051, 1992.
the cation conductance, and urea decreases the Smase activity. 18. Duranton C, Huber SM, and Lang F. Oxidation induces a Cl⫺-depen-
Thus, at least in erythrocytes, both extraordinary Cl⫺ and urea dent cation conductance in human red blood cells. J Physiol 539: 847–
concentrations, as they prevail in renal medulla, partially ab- 855, 2002.
rogate the proapoptotic effects of hyperosmotic shock. The 19. Eda S and Sherman IW. Cytoadherence of malaria-infected red blood
present observations in erythrocytes may thus shed light on the cells involves exposure of phosphatidylserine. Cell Physiol Biochem 12:
373–384, 2002.
complex interplay of NaCl and urea in the regulation of
20. Fadok VA, Bratton DL, Rose DM, Pearson A, Ezekewitz RA, and
medullary kidney cell apoptosis during osmotic shock. Henson PM. A receptor for phosphatidylserine-specific clearance of
ACKNOWLEDGMENTS apoptotic cells. Nature 405: 85–90, 2000.
21. Gamper N, Huber SM, Badawi K, and Lang F. Cell volume-sensitive
We acknowledge the technical assistance of E. Faber and the meticulous sodium channels upregulated by glucocorticoids in U937 macrophages.
preparation of the manuscript by L. Subasic and T. Loch. Pflügers Arch 441: 281–286, 2000.
22. Green DR and Reed JC. Mitochondria and apoptosis. Science 281:
GRANTS
1309 –1312, 1998.
This study was supported by Deutsche Forschungsgemeinschaft Grant Nos. 23. Gulbins E, Bissonnette R, Mahboubi A, Martin S, Nishioka W,
La 315/4-3, La 315/6-1, and La 315/11-1, Bundesministerium für Bildung, Brunner T, Baier G, Baier-Bitterlich G, Byrd C, and Lang F. FAS-
Wissenschaft, Forschung und Technologie (Center for Interdisciplinary Clin- induced apoptosis is mediated via a ceramide-initiated RAS signaling
ical Research) Grant 01 KS 9602, and the Biomed program of the European pathway. Immunity 2: 341–351, 1995.
Union (BMH4-CT96 – 0602). 24. Gulbins E, Jekle A, Ferlinz K, Grassme H, and Lang F. Physiology of
apoptosis. Am J Physiol Renal Physiol 279: F605–F615, 2000.
REFERENCES
25. Gulbins E, Szabo I, Baltzer K, and Lang F. Ceramide-induced inhibi-
1. Agil A and Sadrzadeh SM. Hydroxy-urea protects erythrocytes against tion of T lymphocyte voltage-gated potassium channel is mediated by
oxidative damage. Redox Rep 5: 29 –34, 2000. tyrosine kinases. Proc Natl Acad Sci USA 94: 7661–7666, 1997.

AJP-Renal Physiol • VOL 286 • JUNE 2004 • www.ajprenal.org


INHIBITION OF ERYTHROCYTE “APOPTOSIS” BY UREA AND CL⫺ F1053
26. Huber SM, Gamper N, and Lang F. Chloride conductance and volume- 45. Michea L, Ferguson DR, Peters EM, Andrews PM, Kirby MR, and
regulatory nonselective cation conductance in human red blood cell Burg MB. Cell cycle delay and apoptosis are induced by high salt and
ghosts. Pflügers Arch 441: 551–558, 2001. urea in renal medullary cells. Am J Physiol Renal Physiol 278: F209 –
27. Jarvis WD, Kolesnick RN, Fornari FA, Traylor RS, Gewirtz DA, and F218, 2000.
Grant S. Induction of apoptotic DNA damage and cell death by activation 46. Raines MA, Kolesnick RN, and Golde DW. Sphingomyelinase and
of the sphingomyelin pathway. Proc Natl Acad Sci USA 91: 73–77, 1994. ceramide activate mitogen-activated protein kinase in myeloid HL-60
28. Kaji DM and Gasson C. Urea activation of K-Cl transport in human cells. J Biol Chem 268: 14572–14575, 1993.
erythrocytes. Am J Physiol Cell Physiol 268: C1018 –C1025, 1995. 47. Roger F, Martin PY, Rousselot M, Favre H, and Feraille E. Cell
29. Kaji DM, Lim J, Shilkoff W, and Zaidi W. Urea inhibits the Na-K pump shrinkage triggers the activation of mitogen-activated protein kinases by
in human erythrocytes. J Membr Biol 165: 125–131, 1998. hypertonicity in the rat kidney medullary thick ascending limb of the
30. Koch J and Korbmacher C. Osmotic shrinkage activates nonselective Henle’s loop. Requirement of p38 kinase for the regulatory volume
cation (NSC) channels in various cell types. J Membr Biol 168: 131–139, increase response. J Biol Chem 274: 34103–34110, 1999.
1999. 48. Rosette C and Karin M. Ultraviolet light and osmotic stress: activation
31. Koopman G, Reutelingsperger CP, Kuijten GA, Keehnen RM, Pals of the JNK cascade through multiple growth factor and cytokine receptors.
ST, and van Oers MH. Annexin V for flow cytometric detection of Science 274: 1194 –1197, 1996.
phosphatidylserine expression on B cells undergoing apoptosis. Blood 84: 49. Roy DR, Layton HE, and Jamison RL. Countercurrent mechanism and
1415–1420, 1994. Ist regulation. In: The Kidney. Physiol and Pathophysiol, edited by Seldin
32. Lang F, Busch GL, Ritter M, Völkl H, Waldegger S, Gulbins E, and DW and Gibisch G. New York: Raven, 1992, p. 1649 –1692.

Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on May 11, 2017


Häussinger D. Functional significance of cell volume regulatory mecha- 50. Speake PF and Gibson JS. Urea-stimulated K-Cl cotransport in equine
nisms. Physiol Rev 78: 247–306, 1998. red blood cells. Pflügers Arch 434: 104 –112, 1997.
33. Lang KS, Duranton C, Poehlmann H, Myssina S, Bauer C, Lang F, 51. Tamura H, Noto M, Kinoshita K, Ohkuma S, and Ikezawa H. Inhi-
Wieder T, and Huber SM. Cation channels trigger apoptotic death of bition of NGF-induced neurite outgrowth of PC12 cells by Bacillus cereus
erythrocytes. Cell Death Differ 10: 249 –256, 2003. sphingomyelinase, a bacterial hemolysin. Toxicon 32: 629 – 633, 1994.
34. Lang KS, Myssina S, Tanneur V, Wieder T, Huber SM, Lang F, and 52. Tian W, Boss GR, and Cohen DM. Ras signaling in the inner medullary
Duranton C. Inhibition of erythrocyte cation channels and apoptosis by cell response to urea and NaCl. Am J Physiol Cell Physiol 278: C372–
ethylisopropylamiloride. Naunyn Schmiedebergs Arch Pharmacol 367: C380, 2000.
53. Tian W and Cohen DM. Signaling and gene regulation by urea in cells
391–396, 2003.
of the mammalian kidney medulla. Comp Biochem Physiol A 130: 429 –
35. Lang KS, Roll B, Myssina S, Schittenhelm M, Scheel-Walter HG,
436, 2001.
Kanz L, Fritz J, Lang F, Huber SM, and Wieder T. Enhanced
54. Volk T, Fromter E, and Korbmacher C. Hypertonicity activates non-
erythrocyte apoptosis in sickle cell anemia, thalassemia and glucose-6-
selective cation channels in mouse cortical collecting duct cells. Proc Natl
phosphate dehydrogenase deficiency. Cell Physiol Biochem 12: 365–372,
Acad Sci USA 92: 8478 – 8482, 1995.
2002.
55. Wehner F, Böhmer C, Heinzinger H, van den BF, and Tinel H. The
36. Lang KS, Sandu C, Myssina S, Lang PA, Lang F, Huber SM, and hypertonicity-induced Na(⫹) conductance of rat hepatocytes: physiolog-
Wieder T. Involvment of ceramide in hyperosmotic-shock-induced death ical significance and molecular correlate. Cell Physiol Biochem 10: 335–
of erythrocytes. Cell Death Differ 11: 231–243, 2004 340, 2000.
37. Lang KS, Weigert C, Braedel S, Fillon S, Palmada M, Schleicher E, 56. Wehner F, Sauer H, and Kinne RK. Hypertonic stress increases the
Rammensee HG, and Lang F. Inhibition of interferon-gamma expression Na⫹ conductance of rat hepatocytes in primary culture. J Gen Physiol
by osmotic shrinkage of peripheral blood lymphocytes. Am J Physiol Cell 105: 507–535, 1995.
Physiol 284: C200 –C208, 2003. 57. Wieder T, Orfanos CE, and Geilen CC. Induction of ceramide-mediated
38. Lepple-Wienhues A, Belka C, Laun T, Jekle A, Walter B, Wieland U, apoptosis by the anticancer phospholipid analog, hexadecylphosphocho-
Welz M, Heil L, Kun J, Busch G, Weller M, Bamberg M, Gulbins E, line. J Biol Chem 273: 11025–11031, 1998.
and Lang F. Stimulation of CD95 (Fas) blocks T lymphocyte calcium 58. Williamson P, Christie A, Kohlin T, Schlegel RA, Comfurius P,
channels through sphingomyelinase and sphingolipids. Proc Natl Acad Sci Harmsma M, Zwaal RF, and Bevers EM. Phospholipid scramblase
USA 96: 13795–13800, 1999. activation pathways in lymphocytes. Biochemistry 40: 8065– 8072, 2001.
39. Lim J, Gasson C, and Kaji DM. Urea inhibits NaK2Cl cotransport in 59. Woon LA, Holland JW, Kable EP, and Roufogalis BD. Ca2⫹ sensi-
human erythrocytes. J Clin Invest 96: 2126 –2132, 1995. tivity of phospholipid scrambling in human red cell ghosts. Cell Calcium
40. Maeno E, Ishizaki Y, Kanaseki T, Hazama A, and Okada Y. Normo- 25: 313–320, 1999.
tonic cell shrinkage because of disordered volume regulation is an early 60. Zade-Oppen AM, Schooler JM Jr, Cook P, and Tosteson DC. Effect of
prerequisite to apoptosis. Proc Natl Acad Sci USA 97: 9487–9492, 2000. membrane potential and internal pH on active sodium-potassium transport
41. Martial S, Olives B, Abrami L, Couriaud C, Bailly P, You G, Hediger and on ATP content in high-potassium sheep erythrocytes. Biochim
MA, Cartron JP, Ripoche P, and Rousselet G. Functional differentia- Biophys Acta 555: 285–298, 1979.
tion of the human red blood cell and kidney urea transporters. Am J 61. Zhang Z, Tian W, and Cohen DM. Urea protects from the proapoptotic
Physiol Renal Fluid Electrolyte Physiol 271: F1264 –F1268, 1996. effect of NaCl in renal medullary cells. Am J Physiol Renal Physiol 279:
42. Mason J. The Pathophysiology of ischaemic acute renal failure. Renal F345–F352, 2000.
Physiol 9: 129 –147, 1986. 62. Zhang Z, Yang XY, Soltoff SP, and Cohen DM. PI3K signaling in the
43. Mathias S, Younes A, Kan CC, Orlow I, Joseph C, and Kolesnick RN. murine kidney inner medullary cell response to urea. Am J Physiol Renal
Activation of the sphingomyelin signaling pathway in intact EL4 cells and Physiol 278: F155–F164, 2000.
in a cell-free system by IL-1 beta. Science 259: 519 –522, 1993. 63. Zhou Q, Zhao J, Wiedmer T, and Sims PJ. Normal hemostasis but
44. May A and Huehns ER. The effect of urea on sickling. Br J Haematol 30: defective hematopoietic response to growth factors in mice deficient in
21–29, 1975. phospholipid scramblase 1. Blood 99: 4030 – 4038, 2002.

AJP-Renal Physiol • VOL 286 • JUNE 2004 • www.ajprenal.org

View publication stats

You might also like