1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, and 2 Vascular Biology Unit, Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Tumor necrosis factor
(TNF)- is released in acute inflammatory lung syndromes linked to
the extensive vascular dysfunction associated with increased
permeability and endothelial cell apoptosis. TNF-
induced
significant decreases in transcellular electrical resistance across
pulmonary endothelial cell monolayers, reflecting vascular barrier
dysfunction (beginning at 4 h and persisting for 48 h).
TNF-
also triggered endothelial cell apoptosis beginning at
4 h, which was attenuated by the caspase inhibitor
Z-Val-Ala-Asp-fluoromethylketone. Exploring the involvement
of the actomyosin cytoskeleton in these important endothelial cell
responses, we determined that TNF-
significantly increased myosin
light chain (MLC) phosphorylation, with prominent stress fiber and
paracellular gap formation, which paralleled the onset of decreases in
transcellular electrical resistance and enhanced apoptosis.
Reductions in MLC phosphorylation by the inhibition of either MLC
kinase (ML-7, cholera toxin) or Rho kinase (Y-27632) dramatically
attenuated TNF-
-induced stress fiber formation, indexes of
apoptosis, and caspase-8 activity but not TNF-
-induced
barrier dysfunction. These studies indicate a central role for the
endothelial cell cytoskeleton in TNF-
-mediated apoptosis,
whereas TNF-
-induced vascular permeability appears to evolve
independently of contractile tension generation.
caspases; cytoskeleton; acute lung injury; permeability; Rho
kinase; tumor necrosis factor-
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
ACUTE LUNG INJURY is a common cause of respiratory failure and is characterized by physiological dysfunction attributed to increases in vascular endothelial cell permeability and pulmonary edema formation. Vascular barrier dysfunction in this setting results from endothelial cell activation, leading to cell shape changes and the formation of paracellular gaps (41, 42). The cytoskeletal microfilament proteins actin and myosin constitute 16% of the total endothelial cell protein and exert a critical role in determining cell shape and cellular migration (angiogenesis, wound repair) and in the regulation of endothelial monolayer permeability or barrier function in multiple models (15). The molecular motor that drives nonmuscle actin cytoskeletal rearrangement is the myosin II-associated ATPase, which generates mechanical force by promoting the translational cross-bridge movement of myosin heads across the actin fibers. This process is regulated by the phosphorylation of myosin light chain (MLC) on Ser19 and Thr18 residues catalyzed by the Ca2+/calmodulin-dependent MLC kinase (MLCK). MLC phosphorylation triggers myosin ATPase activity and actin polymerization and is essential to smooth muscle and nonmuscle tension development. Endothelial cell MLCK (214 kDa) is significantly larger than the smooth muscle MLCK isoform (130-160 kDa), although >95% homology exists between the COOH-terminal portions of the endothelial cell and smooth muscle MLCK isoforms. The 922-amino acid NH2 terminus of the endothelial cell MLCK, however, constitutes a completely novel sequence not shared by the smooth muscle MLCK isoform (64). Several distinct endothelial cell MLCK variants, characterized by several specific deletions of nucleotides formed by alternative splicing of a unique transcribed mRNA precursor, have been described (38, 64) and are differentially distributed in various human tissues and cells (38). Endothelial cell MLCK triggers force development after stimulation with rapid activating agonists such as thrombin (13), and this activity is intimately involved in vascular inflammatory responses. For example, inhibition of endothelial cell MLCK activity reduces neutrophil influx into tissues (17) and exerts a marked barrier protective effect in models of ischemia-reperfusion injury (33) and after treatment with vasoactive agonists (13, 55).
Tumor necrosis factor (TNF)- is a proinflammatory cytokine produced
by activated leukocytes and endothelial cells, resulting in
upregulation of endothelial adhesion molecules, alterations in
endothelial cell permeability, and increased edema in isolated perfused
lungs (21, 27). Increased TNF-
levels are found in
bronchoalveolar lavage fluid from patients with acute respiratory distress syndrome (29, 46, 62) and contribute
significantly to the organ dysfunction observed in acute inflammatory
syndromes. Although the mechanisms by which TNF-
increases vascular
barrier dysfunction are not well understood, there is evidence to
support an active role for actin rearrangement (20),
potentially evolving in a MLCK-dependent manner (43).
TNF-
is also a well-recognized potent stimulus for programmed cell
suicide or apoptosis, an important feature of inflammatory
processes including acute lung injury (24, 26, 65, 66).
TNF-
induces endothelial cell apoptosis in vivo and in
vitro, and direct administration of TNF-
produces widespread
endothelial cell apoptosis, predominantly in the pulmonary vascular bed, as well as pulmonary edema (24, 53, 54, 56, 60).
Although information is limited, there is increasing appreciation that
the microfilamentous cytoskeleton may be intrinsically involved in the
apoptosis process by regulating intracellular signaling or by
transmitting death messages to downstream effectors. Cytoskeletal
components such as actin, the actin binding protein gelsolin, and focal
adhesion kinase are all cleaved by caspases (1, 19, 32, 36, 39,
40, 45), and actin polymerization is required for the initiation
of membrane blebbing, a recognized feature of apoptotic cell death
(37). It has been suggested that MLCK may be required for
membrane blebbing in specific models of apoptosis induced by
serum deprivation (12, 47) and, in a single report
(69), TNF--mediated DNA fragmentation in the tumor cell
line U937 was reduced by MLCK inhibition. In this study, we examined
the hypothesis that the microfilament-based cytoskeleton is a critical
participant in TNF-
-induced endothelial cell permeability and
apoptosis. Our results indicate that MLCK and Rho GTPases, two
effectors that work in tandem to increase MLC phosphorylation, are
significantly involved in TNF-
-induced apoptosis and
actomyosin rearrangement but do not appear to be critical participants
in TNF-
-induced endothelial cell barrier dysfunction. Furthermore, inhibition of the execution phase of apoptosis did not alter
the TNF-
-mediated increase in permeability. Elucidation of
cytoskeletal participation in cell survival or apoptosis
signaling may provide important clues regarding the role of endothelial
cell apoptosis in the pathogenesis of acute lung injury.
![]() |
METHODS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Cell culture conditions and reagents.
For most experiments, the bovine pulmonary artery endothelial cells
(BPAECs) were obtained either at the 16th passage (CCL 209; American
Type Culture Collection, Manassas, VA) or at the 2nd passage (caspase-8
activity assay, diphospho-MLC-specific immunoblotting experiments; Cell
Systems, Kirkland, WA). The cells were maintained in complete culture
medium consisting of 20% bovine serum, 17 µg/ml of endothelial cell
growth supplement (Upstate Biotechnology, Lake Placid, NY), and
100 U/ml of penicillin-streptomycin (GIBCO BRL) at 37°C in an
atmosphere of 5% CO2 and 95% air. Caspase inhibitor I [Z-Val-Ala-Asp (ZVAD)-fluoromethylketone
(fmk)], caspase-3 inhibitor I
[N-acetyl-Asp-Glu-Val-Asp-aldehyde
(Ac-DEVD-CHO)], caspase-8 inhibitor II
[Z-Ile-Glu(OMe)-Thr-Asp(OMe)
(Z-IETD)-fmk], 1-(5-iodonapthalene-1-sulfonyl)homopiperazine hydrochloride
(ML-7), and cholera toxin (type Inaba 569B) were from
Calbiochem-Novabiochem (La Jolla, CA). The Rho-associated protein
kinase inhibitor Y-27632 was obtained from Upstate Biotechnology.
Thrombin, TNF- (with a biological activity of 2 × 107 U/mg), and monoclonal anti-human TNF-
antibody
(clone TA-31) were from Sigma-Aldrich (St. Louis, MO). Texas Red-X
phalloidin was purchased from Molecular Probes (Eugene, OR).
Anti-diphosphorylated MLC antibody generation. Antisera production and peptide syntheses were performed by Sigma-Genosys (The Woodlands, TX). Phospho-specific MLC antibodies were generated by injecting rabbits with the KLH-conjugated phosphorylated peptides KKRPQRATS[p]NVFA (monophospho-specific antibodies) or KKRPQAT[p]S[p]NVFA (diphospho-specific MLC antibodies), where [p] indicates that the residue to the left has been chemically phosphorylated. These peptides encompass MLC phosphorylation sites Thr18 and Ser19. Phospho-specific antibodies were obtained by a two-step affinity purification procedure. The first step involved removing antibodies generated against the nonphosphorylated region of the peptide by passing the crude antisera through a column containing nonphosphorylated peptide (KKRPQRATSNVFA) conjugated to Sepharose. The "flow-through" from this column showed a large enrichment for specific antibodies to monophospho-MLC and diphospho-MLC. This was then affinity purified in a second step by passing it over a column containing either the mono- or diphosphorylated peptides and collecting the eluant.
Apoptosis assays: endothelial cell annexin V staining. BPAECs were stained with annexin V and propidium iodide with the ApoAlert annexin V apoptosis kit (Clontech Laboratories, Palo Alto, CA). Detached cells were recovered by centrifugation, resuspended in the binding buffer, and added back to the tissue culture plate before being stained. Apoptotic cells were detected by fluorescence microscopy with an Eclipse TE300 inverted microscope (Nikon, Melville, NY). Apoptotic cells stained green at the plasma membrane. Necrotic or late apoptotic cells took up propidium iodide and stained red. A quantitative estimation was made by counting apoptotic cells relative to the total number of cells seen within the counted fields (with the use of bright-field microscopy). The apoptosis index reported reflects the number of annexin-stained cells divided by the total number of cells on the monolayer within the same microscopic field and is expressed relative to the number obtained in unstimulated control conditions (multiple of increase).
DNA electrophoresis. Both DNA laddering and nucleosome ELISA methods have high specificity and recognize a more downstream apoptotic event, internucleosomal DNA cleavage. Genomic DNA was obtained from cultured bovine endothelial cells after specific interventions with the Puregene DNA isolation kit (Gentra Systems, Minneapolis, MN), following the manufacturer's instructions. Briefly, cells were lysed with a solution containing SDS and Tris-EDTA. RNase was added, and the proteins were precipitated with ammonium acetate solution. DNA was precipitated with isopropanol, resuspended in Tris-EDTA, and electrophoresed on a 1.5% agarose-ethidium bromide gel at 6 V/cm. The resulting gel was then photographed under ultraviolet luminescence with a Polaroid camera. Apoptosis was recognized by the presence of discrete bands of DNA that migrated in the range of 100-1,000 bp (laddering), whereas necrosis was characterized by the appearance of a "smear" of DNA. Commercially obtained DNA fragments of 1-kb and 100-bp size were used as markers.
Nucleosome ELISA. This method has the advantage of being easily quantifiable. Quantitation of apoptotic endothelial cells was obtained with a nucleosome ELISA kit (Oncogene Research Products, Cambridge, MA) following the manufacturer's protocol. In these experiments, endothelial cells were lysed, and the supernatant was loaded onto precoated DNA-binding protein wells. The nucleosomes were detected with anti-histone H3-biotinylated antibody followed by streptavidin-horseradish peroxidase, with absorbance (450 nm) compared with lyophilized standards with designated nucleosome unit values.
Caspase activity assay. Caspase-8 activity was assayed with the ApoAlert caspase-8 colorimetric assay kit from Clontech used following the manufacturer's instructions. Cells were lysed and centrifuged, and the supernatant was assayed for protease activity with the specific chromogenic substrate Ac-IETD-p-nitroanilide (pNA), with optical density measurements at 400 nm taken every 30 min for up to 180 min in a Vmax microplate reader (Molecular Devices, Sunnyvale, CA). The slopes of the curves obtained were normalized to those of blank samples (buffer and lysis buffer only). Caspase activity units were calculated by dividing the values obtained by the slope of a standard curve of absorbance of the chromogen alone (pNA).
Endothelial monolayer resistance measurements.
The electrical resistance of BPAEC monolayers was measured with
the electrical cell impedance sensor technique that our laboratory (16) has previously described. In this system
(Applied Biophysics, Troy, NY), endothelial cells were cultured on a
small gold electrode (104 cm2) in complete
medium. The endothelial monolayers act as insulating particles, and the
total resistance across the monolayers is composed of the resistance
between the ventral cell surface and the electrode and the resistance
between cells. A 4,000-Hz AC signal with 1-V amplitude through a
1-M
resistor created an approximate constant-current source. The lock-in amplifier attached to the electrodes detected changes in both magnitude and phase of the voltage that appeared across
the endothelial cell and was controlled by an IBM-compatible personal
computer that was used both for data accumulation and processing.
Transcellular electrical resistance (TER) increased immediately after
cell attachment and achieved a steady state when endothelial cells
became confluent. Thus experiments were conducted after the electrical
resistance achieved a steady state. Resistance data were normalized to
the initial voltage and plotted as a normalized TER. Only wells in
which the TER achieved >5,000
were utilized.
MLC immunoprecipitation.
For immunoprecipitation under denaturing conditions, confluent
endothelial cell monolayers in 60-mm tissue culture dishes were labeled
with [32P]orthophosphate (0.5 mCi/plate) for 2.5 h
in phosphate-free DMEM (Sigma) with 1% serum, followed by stimulation
with either vehicle alone, TNF-, or thrombin. The stimuli were then
removed, and the monolayers were rinsed twice with 2 ml of medium,
further rinsed with 2 ml of PBS, and scraped into 200 µl of
SDS-denaturing stop solution (PBS, pH 7.4, 1 mM EDTA, 1 mM EGTA, 50 mM
NaF, 10 mM sodium pyrophosphate, 0.2 mM orthovanadate, 1%
SDS, and 14 mM
-mercaptoethanol). The homogenate was
prepared by passing the cell suspension through a 16-gauge needle
several times. Homogenates were heat treated at 110°C for 5 min,
diluted 1:10 with 900 µl of PBS, and incubated with 50 ml of 10%
Pansorbin suspension (formalin-hardened and heat-killed Cowan 1 strain
Staphylococcus aureus cells; Calbiochem) for 30 min at room
temperature. Samples were clarified by microcentrifugation for 5 min,
and supernatants were incubated first with 20 µl of anti-MLC antibody
(Biodesign International, Kennebunk, ME) for 60 min at room temperature
or overnight at 4°C and then with 50 µl of 10% Pansorbin
suspension for 60 min at room temperature. Immunocomplexes were
pelleted by microcentrifugation for 5 min, washed three times with 1 ml
of PBS, solubilized in 100 µl of boiled 2× SDS-Laemmli sample
buffer, and then were separated from the Pansorbin beads by
microcentrifugation and subjected to SDS electrophoresis. After
electrophoresis, the proteins were transferred to nitrocellulose
membranes, and 32P signals were detected by autoradiography
at
70°C. The relative intensities of the 32P-labeled
MLC were quantified by scanning densitometry.
Actin and diphosphorylated MLC immunofluorescence. BPAECs were cultured in 12-well dishes on coverslips coated with gelatin until a confluent monolayer was achieved. After exposure to experimental conditions, endothelial cell monolayers were fixed in 3.7% formaldehyde and were permeabilized with 0.25% Triton X-100. After being stained with Texas Red-X phalloidin (1:200), the coverslips were mounted on slides and examined under oil immersion (×60 magnification) with an Eclipse TE300 inverted microscope (Nikon). Actin was visualized by Texas Red-X phalloidin staining (Molecular Probes) for 1 h at room temperature and stained red. After exposure to experimental conditions, the coverslips were fixed in 3.7% formaldehyde and permeabilized with 0.25% Triton X-100. This method enabled the examination of endothelial cell morphology (cellular rounding, shrinkage), intercellular gap formation in confluent monolayers, and intracellular actin filament reorganization (stress fiber formation, cortical or perinuclear actin organization). Staining for MLC was performed in a similar manner with a primary antibody that was immunoreactive with diphosphorylated MLC. After three washes with PBS, the monolayers were incubated with an appropriate secondary antibody conjugated to immunofluorescent dyes (Alexa 488 for green fluorescence or Alexa 546 for red fluorescence) for 1 h at room temperature. After three washes in PBS, the coverslips were mounted and analyzed with a Nikon video imaging system consisting of a phase-contrast inverted microscope connected to a digital camera connected to an image processor, and the images were recorded and saved in an Adobe Photoshop 4 program on a Pentium II PC.
Western immunoblotting. Endothelial cell proteins were separated by SDS-PAGE, transferred to nitrocellulose membranes, and immunoblotted for 2 h with a polyclonal antibody directed at diphosphorylated MLC. This was followed by the addition of the appropriate horseradish peroxidase-conjugated secondary antibody (1:10,000). The reaction was visualized by enhanced chemiluminescence and autoradiography (Amersham) according to the manufacturer's instructions. The quantification of the immunoreactive proteins present was made with a Bio-Rad GL-670 scanning densitometer.
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Effects of TNF- on TER.
Our initial studies examined TNF-
-induced changes in endothelial
cell permeability utilizing the electrical cell impedance sensor
technique in which endothelial cells are grown to confluence on gold
microelectrodes and TER is measured and expressed as normalized resistance to the initial measured voltage. TNF-
caused significant time- and dose-dependent decreases in TER beginning at 4-5 h of exposure and reaching a maximum decline at 10 h, with changes persisting for 24-48 h (Fig. 1). The
dose-dependent effects of TNF-
on endothelial cell barrier function
plateaued at 100 ng/ml of TNF-
and were specific to TNF-
because
an anti-TNF-
monoclonal antibody completely abolished the
TNF-
-induced changes in TER (Fig. 1). In contrast to the delayed
TNF-
-induced decreases in TER, thrombin, a proinflammatory mediator
our laboratory (13) has previously shown to cause
significant endothelial cell barrier dysfunction and vascular
permeability, produced a prompt and immediate decline (onset in
minutes) in TER, with subsequent recovery (Fig. 1).
|
Effect of TNF- on endothelial cell
MLC phosphorylation and actin rearrangement.
We next examined whether the myosin-driven endothelial cell contractile
apparatus is involved in TNF-
-induced endothelial cell activation
using several complementary strategies to test this hypothesis. We
first immunoprecipitated MLCs from 32P-labeled endothelial
cells with anti-MLC antibody at specified periods (5 min, 30 min, and
1 h) after TNF-
challenge. These studies demonstrated an almost
threefold increase in MLC phosphorylation at 1 h compared
with baseline values (Fig.
2A). The TNF-
-induced increase in MLC phosphorylation was similar to that in the positive control, diperoxovanadate, and was sustained up to 6 h as
demonstrated by diphospho-MLC-specific immunoblotting (Fig.
2B). TNF-
-induced increases in MLC phosphorylation were
also evaluated and confirmed by immunofluorescence microscopy with
anti-diphosphorylated specific MLC antibody, where a significant
increase in fluorescence at 1 (data not shown) and 4 h (Fig.
2C) was demonstrated after TNF-
. Pretreatment (30 min)
with the selective MLCK inhibitor ML-7 (48) inhibited
TNF-
-induced MLC phosphorylation in both an in vitro MLC
phosphorylation assay with purified recombinant human endothelial cell
MLCK and recombinant MLC (data not shown) and in vivo (Fig. 2B), suggesting that TNF-
-induced MLC phosphorylation is
catalyzed, at least in part, by MLCK. Rho kinase also participates in
MLC phosphorylation regulation by phosphorylation of the regulatory subunits of the myosin-associated phosphatase (8).
Endothelial cell pretreatment with Y-27632, a Rho kinase inhibitor
(4), prevented TNF-
-induced MLC phosphorylation (Fig.
2, B and C), implicating the involvement of Rho
kinase activation in TNF-
-induced increases in MLC phosphorylation.
|
|
Effect of MLCK and Rho kinase
inhibition on TNF--induced endothelial cell barrier
dysfunction.
We next examined whether MLCK and Rho kinase were involved in
TNF-
-induced endothelial cell barrier dysfunction. Endothelial cell
monolayers grown on gold microelectrodes were treated with either the
MLCK inhibitor ML-7 or the Rho kinase inhibitor Y-27632 before TNF-
challenge. Despite significant evidence of MLCK activation, neither
inhibition of MLCK (2-100 µM; Fig.
4A) nor Rho kinase (data not
shown) altered TNF-
-mediated declines in TER. These data suggest
that neither MLCK nor Rho kinase activation is a critical participant
in the permeability response elicited by TNF-
. Identical results
were obtained with the additional MLCK inhibitor ML-9 (data not shown)
as well as with cholera toxin (Fig. 4B), which, via
cAMP-dependent PKA activation, reduces MLCK activity and MLC
phosphorylation (13) and is a remarkably effective barrier-protective agent in multiple models of vascular permeability (50, 59). These data suggest that despite significant
MLCK- and Rho kinase-dependent MLC phosphorylation and active
cytoskeletal rearrangement, these events are not central to the
evolution of TNF-
-induced endothelial barrier dysfunction.
|
TNF--induced apoptosis in
BPAECs.
We next defined the in vitro conditions required for the
well-recognized ability of TNF-
to increase endothelial cell
apoptosis. Complementary assays including DNA electrophoresis,
annexin V immunofluorescence microscopy, and nucleosomal ELISA were
used. TNF-
(at both 20 and 100 ng/ml) induced significant increases in the number of annexin-positive and propidium iodide-negative endothelial cells beginning as early as 4 h after
exposure (Fig. 5A). The higher
concentration of TNF-
(100 ng/ml) also increased the number of
propidium iodide-positive cells (90-fold compared with control value),
reflecting either necrotic or late apoptotic cells. A concentration
of 20 ng/ml of TNF-
was therefore used in subsequent experiments
involving apoptosis. To confirm the early membrane-related
changes associated with apoptosis, we performed DNA
electrophoresis and nucleosomal ELISA assays, which detect later stages
of apoptosis (i.e., internucleosomal DNA cleavage). The results
showed a strong time-dependent increase in the apoptotic index, up
to 96% at 20 h by ELISA (Fig. 5B), with the detection of typical DNA laddering at 12 h by DNA electrophoresis (Fig. 6B). As with changes in
electrical resistance, the effects of TNF-
on apoptosis were
specific because the addition of a monoclonal anti-TNF-
antibody
completely abolished TNF-
-induced cell death (data not shown).
Predictably, pretreatment with the irreversible general caspase
inhibitor ZVAD-fmk (100 µM for 40 min) also effectively inhibited
TNF-
-induced endothelial cell apoptosis (Fig. 6).
|
|
Effect of MLCK and Rho kinase
inhibition on TNF--induced apoptosis.
We next tested the hypothesis that the apoptotic process requires
active MLCK-driven microfilamentous cytoskeletal
rearrangement. Inhibition of MLCK, either directly by ML-7 or
indirectly by cholera toxin (2 µg/ml for 1 h) (51),
significantly reduced endothelial cell apoptosis triggered by
TNF-
(Fig. 7). The inhibitory action of the cell-permeable ML-7 was particularly dramatic when assessed with
the sensitive annexin assay at 4 h (Fig. 7), whereas cholera toxin-mediated protection was more effective at later times, perhaps reflecting the inherently slower inhibition of MLCK by cholera toxin-elicited ADP ribosylation of Gs- and subsequent
cAMP-mediated PKA activation (50) (Fig. 7). To further
characterize the participation of the MLC phosphorylation-dependent
cytoskeletal changes in the apoptotic process, we next investigated
the effect these changes may have on the activation of the caspase
cascade. Inhibition of MLCK by ML-7 (10 µM) resulted in a 54%
attenuation of the TNF-
-induced increase (sixfold) of caspase-8
activity (Fig. 8), an upstream component
of the caspase cascade, as demonstrated by cleavage of a specific
chromogenic substrate, Ac-IETD-pNA. Given the temporal similarities in
both the increases in permeability and the onset of apoptosis
evoked by TNF-
, we next examined the potential role of
apoptosis in endothelial cell permeability by inhibiting
apoptosis with ZVAD-fmk and evaluating TNF-
-induced
reductions in the TER. As seen in Fig. 9,
ZVAD-fmk failed to significantly alter the onset of TNF-
-mediated
permeability. Similar results were obtained with Ac-DEVD-CHO,
a cell-permeable, reversible general caspase inhibitor, and with
Z-IETD-fmk, a more specific caspase-8 inhibitor (data not
shown).
|
|
|
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
The participation of TNF- in pulmonary pathophysiology is well
recognized, with specific activation of vascular cellular components
resulting in endothelial cell barrier dysfunction (3, 10, 22,
49), transendothelial leukocyte diapedesis into tissues
(23, 63), and marked increases in endothelial cell apoptosis (30, 52-54, 56, 58). In this
report, we have addressed potential endothelial cell
cytoskeleton-dependent mechanisms that may underlie the effects of
TNF-
on vascular barrier dysfunction and endothelial cell
apoptosis. The endothelial cytoskeleton, a key determinant of
cell shape and migration (angiogenesis, wound repair), is responsible
for the dynamic nature of the paracellular junction regulation that is
so vital to models of endothelial permeability (15). Our
data provide several lines of evidence that significant activation of
the endothelial cell contractile apparatus occurs in response to
TNF-
. We noted that TNF-
-induced actin rearrangement and
intercellular gap formation temporally coincided with the
phosphorylation of MLCs (1-2 h) and preceded the onset of both
permeability and apoptosis, suggesting a linkage between the
actin cytoskeletal changes and both apoptosis and the
permeability responses to the cytokine. Consistent with the role of
MLCK in cytoskeletal rearrangement, MLCK inhibition with ML-7 markedly
attenuated TNF-
-induced MLC phosphorylation, cell contraction, and
stress fiber formation. Furthermore, MLCK inhibition significantly
attenuated TNF-
-mediated endothelial apoptosis. Unlike
models of vascular permeability after ischemia-reperfusion injury or treatment with thrombin, MLCK inhibition did not prevent the
delayed permeability response that ensues after TNF-
, reflecting the
mechanistic specificity of different proinflammatory and edemagenic agents.
Our data are consistent with the evolving concept that the
actomyosin-based cytoskeleton is a critical participant in
TNF--induced endothelial cell activation, particularly with respect
to programmed cell death. TNF-
produces active actin cytoskeletal
rearrangement in endothelium (Fig. 3) associated with an increase in
the G-actin pool (20), stress fiber formation, and
intercellular gap formation involving Rho-related GTPases (3,
67). The cytoskeletal rearrangement in response to TNF-
is
associated with MLC phosphorylation mediated by both MLCK and Rho
kinase (Fig. 2). These results are in agreement with the work of Medina
et al. (43), who previously observed a 23-kDa protein
comigrating with MLC standards after TNF-
stimulation (100 ng/ml).
It is well recognized that Rho kinases increase MLC phosphorylation via
inhibition of the regulatory subunit of the myosin-specific
phosphatase, whereas Rho inhibition decreases MLC phosphorylation
(18). We have shown that biochemical inhibition of Rho
kinase and MLCK reduced both MLC phosphorylation and actin filament
rearrangement in response to TNF-
(Figs. 2 and 3), implicating Rho
kinase and MLCK activation as essential steps in the mechanism of
TNF-
-mediated cytoskeletal changes. Interestingly, Rho inactivation by Clostridium botulinum C3 toxin also decreased membrane
blebbing, suggesting that Rho kinase may also participate in bleb
formation (47). The activation of MLCK by TNF-
appears
critical for the execution of the programmed cell death induced by the
cytokine because inhibition of MLC phosphorylation significantly
attenuated TNF-
-induced apoptosis (Fig. 7). This effect
suggests an essential role of microfilament cytoskeleton rearrangement
in the assembly of the intracellular death-activating pathways. Several
authors (32, 40) have detected the cleavage of actin
during programmed cell death, although this remains controversial
(57). Fas-induced damage of actin was associated with
caspase-3-induced cleavage of the actin-severing and -capping protein
gelsolin. Fragmented gelsolin overexpression induces DNA fragmentation,
suggesting an effector role of gelsolin cleavage in apoptotic
morphological changes (36). Destabilization of the actin
cytoskeleton may contribute to the activation of key apoptotic
regulators because cytochalasin D, which potently induces actin
depolymerization, also triggers significant apoptosis (6,
61). Overexpression of the actin monomer binding protein
thymosin-
10 accelerates apoptosis in a manner consistent
with the notion that cytoskeletal rearrangement may be a critical event
in endothelial cell apoptosis (6, 25). The
existence of a direct link between actin depolymerization and DNA
degradation is not known with certainty; however, it has been suggested
that DNase I may reside in the ends of actin filaments, subsequently
becoming liberated by G-actin destruction (32).
The direct involvement of MLC phosphorylation in apoptosis has
also been suggested by studies of membrane blebbing and DNA fragmentation in cultured cells. Huot et al. (28) have
shown that the membrane blebbing during
H2O2-induced endothelial cell (human umbilical
vein) apoptosis is regulated by F-actin reorganization, which,
in turn, is dependent on stress-activated protein kinase/p38 activation
via phosphorylation of heat shock protein-27, an actin polymerization
modulator (28). The actin changes lead to disassembly of
focal adhesions and membrane blebbing. Actin-myosin interaction, regulated by MLCK, proved to be a key factor in membrane blebbing of
PC-12 cells induced by serum withdrawal (47). However, the membrane blebbing and the execution phase of apoptosis may be dissociated, as previously reported (44). Furthermore,
TNF--mediated DNA fragmentation in the tumor cell line U937 was
blocked by a potent inhibitor of MLCK but was unaffected by inhibitors
of cAMP- or cGMP-dependent protein kinases (69).
TNF- fragments form active trimers, which exert systemic effects via
ligation of two classes of TNF-
receptors, 55-kDa receptor type 1 (TNFR1) and 75-kDa receptor type 2 (TNFR2), both of which are found on
the membranes of virtually all nucleated cells. Apoptotic TNF-
signals to endothelial cells are transmitted through TNFR1, one of the
five known apoptosis surface receptors that contains an
intracellular protein motif known as the death domain. Once clustered
at the activated receptor, these proteins relay as yet unknown signals
downstream to a procaspase (procaspase-8), with subsequent activation
of the caspase cascade (caspases 3, 6, and 7) and simultaneous
inhibition of the nuclear factor
B-mediated antiapoptotic
pathways, resulting in execution of the apoptotic program and the
typical morphological changes of cellular shrinkage and apoptotic
body formation. It was recently demonstrated that apoptotic
regulation in myocytes involves a protein, ARC, an apoptosis repressor with a caspase recruitment domain activated by hypoxia or
reactive oxygen species, which regulates apoptosis by acting upstream of caspase activation (35). ARC overexpression
rescues HEK293 cells from apoptosis caused by cotransfection of
the death receptor FAS and TNFR1 as well as the death domain adaptors
TNF receptor-associated death domain and caspase and
receptor-interacting protein adapter with death domain, which
regulate TNFR1-evoked bifurcating pathways, resulting in caspase-8
activation (7). One potential mechanism for MLCK
involvement in TNF-
-induced apoptosis in endothelium may
involve caspase cascade activation facilitated by actomyosin
contractile rearrangement that is critical to the assembly of the
TNF-
death receptor complex. Garcia et al. (14)
previously cloned endothelial cell MLCK and subsequently identified
five splice variants whose exact role in cytoskeletal regulation has
not yet been characterized (38). Endothelial cell MLCK is
highly homologous in the COOH-terminal portion of the enzyme to smooth
muscle MLCK, and recently, two protein kinases having significant
homology to both smooth muscle and endothelial cell MLCK isoforms were
identified and exhibited discrete kinase, calmodulin-binding, and
ankyrin repeat regions and a death domain. One of these kinases is the
DAP kinase, which has been suggested to promote apoptosis
(31). Whether a specific MLCK splice variant participates
in cytokine-stimulated programmed cell death is not yet known, but the
idea provides a potentially intriguing mechanism for isoform-specific
MLCK regulation of endothelial cell apoptosis.
Both apoptosis and increased vascular permeability have been
observed in in vitro and in vivo models of TNF--induced injury, but
their interdependency is not known (24). The onset of
permeability after TNF-
exposure (4-5 h, maximum at 10 h)
is in agreement with previous studies (10, 11, 22, 49)
that relied primarily on the measurement of albumin flux across the
endothelial cell monolayer measured over a 6-h exposure time. We
confirmed that TNF-
induces apoptosis in BPAECs as early as
4 h, which coincides with the onset of permeability; hence, we
evaluated the contribution of apoptotic cell death to
TNF-
-induced endothelial cell permeability by inhibiting the
execution phase of apoptosis. We have demonstrated that the
changes in permeability secondary to TNF-
are not the direct result
of caspase-mediated apoptotic death and, in fact, were quite
impressed that after an initial decline in TER, barrier function
remained stable, maximizing at 10 h, despite continued and
substantial increases in endothelial cell apoptosis. These results suggest the presence of highly efficient cell-cell connections that contribute to the maintained stability of the endothelial monolayer and barrier by preventing paracellular gap formation. One
potential explanation for the dichotomous TNF-
response is that
TNF-
-induced permeability may be completely independent of
apoptosis, involving discrete signaling cascades such as
TNF-
-induced release of reactive oxygen and nitrogen species
(11). Nevertheless, it would appear clear that although
the precise mechanisms of TNF-
-induced endothelial cell barrier
dysfunctions have not yet been elucidated, the rearrangement of the
actin-based cytoskeleton is not the critical determinant of the
TNF-
-mediated endothelial cell barrier dysfunction as is the case,
for example, for specific bioactive agonists such as thrombin. MLC
phosphorylation occurred in response to TNF-
in association with
cytoskeletal changes and temporally before the onset of endothelial
cell barrier dysfunction (Figs. 1 and 2). However, inhibition of MLC
phosphorylation by either MLCK or Rho kinase did not attenuate
TNF-
-induced endothelial cell permeability, indicating that MLCK
activation alone is insufficient to induce barrier dysfunction.
Conversely, the mechanisms by which TNF-
induces endothelial cell
barrier dysfunction may not primarily involve MLCK-regulated
actin-myosin interaction but involve other cytoskeletal components such
as intermediate filaments, microtubules, and adherens junction
proteins, which may also play a role in TNF-
-induced vascular
barrier dysfunction (2, 5, 9, 68). It is thought that
cyclic nucleotide-dependent protein kinases might be important
mediators of the TNF-
-induced barrier dysfunction and that protein
kinase C and calmodulin do not appear to be involved in this response
(34, 70). More recently, however, Ferro and colleagues
(10, 11) have implicated PKC and nitric oxide in at least
the initial (first 4-6 h) phases of TNF-
-induced endothelial
cell permeability.
In summary, we have examined the role of the actin-myosin microfilament
system in TNF--induced endothelial cell apoptosis and
barrier dysfunction. We have shown that MLCK is critical for the
execution of TNF-
-induced apoptosis signal progression in endothelial cells, which is consistent with an active role of the
actin-myosin interaction in the TNF-
-induced apoptotic signaling in endothelial cells. It is possible that the actomyosin contraction is
needed for the spatial changes that favor molecular interactions of
proapoptotic signals. The differential role of MLCK-dependent cytoskeletal changes in TNF-
-induced endothelial cell
apoptosis and permeability, as well as their apparent
dichotomy, underscores the vast complexity of the regulation of the
biological response to this cytokine in inflammatory injury cascades.
![]() |
ACKNOWLEDGEMENTS |
---|
We gratefully acknowledge Steve Durbin, Peiyi Wang, Keri Jacobs, and Lakshmi Natarajan for superb technical assistance and Ellen Reather for manuscript preparation.
![]() |
FOOTNOTES |
---|
This work was supported by National Heart, Lung, and Blood Institute Grants HL-50533, HL-58064, and HL-04396.
Address for reprint requests and other correspondence: J. G. N. Garcia, Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Cir., Rm. 4B.44, Baltimore, MD 21224-6801 (E-mail: drgarcia{at}welch.jhu.edu).
The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Received 31 October 2000; accepted in final form 14 December 2000.
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
1.
Bannerman, DD,
Sathyamoorthy M,
and
Goldblum SE.
Bacterial lipopolysaccharide disrupts endothelial monolayer integrity and survival signaling events through caspase cleavage of adherens junction proteins.
J Biol Chem
273:
35371-35380,
1998
2.
Blum, MS,
Toninelli E,
Anderson JM,
Balda MS,
Zhou J,
O'Donnell L,
Pardi R,
and
Bender JR.
Cytoskeletal rearrangement mediates human microvascular endothelial tight junction modulation by cytokines.
Am J Physiol Heart Circ Physiol
273:
H286-H294,
1997
3.
Brett, J,
Gerlach H,
Nawroth P,
Steinberg S,
Godman G,
and
Stern D.
Tumor necrosis factor/cachectin increases permeability of endothelial cell monolayers by a mechanism involving regulatory G proteins.
J Exp Med
169:
1977-1991,
1989[Abstract].
4.
Carbajal, JM,
Gratrix ML,
Yu CH,
and
Schaeffer RC, Jr.
ROCK mediates thrombin's endothelial barrier dysfunction.
Am J Physiol Cell Physiol
279:
C195-C204,
2000
5.
Curtis, TM,
Rotundo RF,
Vincent PA,
McKeown-Longo PJ,
and
Saba TM.
TNF--induced matrix Fn disruption and decreased endothelial integrity are independent of Fn proteolysis.
Am J Physiol Lung Cell Mol Physiol
275:
L126-L138,
1998
6.
DeMeester, SL,
Cobb JP,
Hotchkiss RS,
Osborne DF,
Karl IE,
Tinsley KW,
and
Buchman TG.
Stress-induced fractal rearrangement of the endothelial cell cytoskeleton causes apoptosis.
Surgery
124:
362-371,
1998[ISI][Medline].
7.
Ekhterae, D,
Lin Z,
Lundberg MS,
Crow MT,
Brosius FC, III,
and
Nunez G.
ARC inhibits cytochrome c release from mitochondria and protects against hypoxia-induced apoptosis in heart-derived H9c2 cells.
Circ Res
85:
e70-e77,
1999
8.
Essler, M,
Amano M,
Kruse HJ,
Kaibuchi K,
Weber PC,
and
Aepfelbacher M.
Thrombin inactivates myosin light chain phosphatase via Rho and its target Rho kinase in human endothelial cells.
J Biol Chem
273:
21867-21874,
1998
9.
Ferrero, E,
Villa A,
Ferrero ME,
Toninelli E,
Bender JR,
Pardi R,
and
Zocchi MR.
Tumor necrosis factor alpha-induced vascular leakage involves PECAM1 phosphorylation.
Cancer Res
56:
3211-3215,
1996[Abstract].
10.
Ferro, TJ,
Gertzberg N,
Selden L,
Neumann P,
and
Johnson A.
Endothelial barrier dysfunction and p42 oxidation induced by TNF- are mediated by nitric oxide.
Am J Physiol Lung Cell Mol Physiol
272:
L979-L988,
1997
11.
Ferro, T,
Neumann P,
Gertzberg N,
Clements R,
and
Johnson A.
Protein kinase C- mediates endothelial barrier dysfunction induced by TNF-
.
Am J Physiol Lung Cell Mol Physiol
278:
L1107-L1117,
2000
12.
Fishkind, DJ,
Cao LG,
and
Wang YL.
Microinjection of the catalytic fragment of myosin light chain kinase into dividing cells: effects on mitosis and cytokinesis.
J Cell Biol
114:
967-975,
1991[Abstract].
13.
Garcia, JG,
Davis HW,
and
Patterson CE.
Regulation of endothelial cell gap formation and barrier dysfunction: role of myosin light chain phosphorylation.
J Cell Physiol
163:
510-522,
1995[ISI][Medline].
14.
Garcia, JG,
Lazar V,
Gilbert-McClain LI,
Gallagher PJ,
and
Verin AD.
Myosin light chain kinase in endothelium: molecular cloning and regulation.
Am J Respir Cell Mol Biol
16:
489-494,
1997[Abstract].
15.
Garcia, JG,
and
Schaphorst KL.
Regulation of endothelial cell gap formation and paracellular permeability.
J Investig Med
43:
117-126,
1995[ISI][Medline].
16.
Garcia, JG,
Schaphorst KL,
Shi S,
Verin AD,
Hart CM,
Callahan KS,
and
Patterson CE.
Mechanisms of ionomycin-induced endothelial cell barrier dysfunction.
Am J Physiol Lung Cell Mol Physiol
273:
L172-L184,
1997
17.
Garcia, JG,
Verin AD,
Herenyiova M,
and
English D.
Adherent neutrophils activate endothelial myosin light chain kinase: role in transendothelial migration.
J Appl Physiol
84:
1817-1821,
1998
18.
Garcia, JG,
Verin AD,
Schaphorst K,
Siddiqui R,
Patterson CE,
Csortos C,
and
Natarajan V.
Regulation of endothelial cell myosin light chain kinase by Rho, cortactin, and p60src.
Am J Physiol Lung Cell Mol Physiol
276:
L989-L998,
1999
19.
Geng, YJ,
Azuma T,
Tang JX,
Hartwig JH,
Muszynski M,
Wu Q,
Libby P,
and
Kwiatkowski DJ.
Caspase-3-induced gelsolin fragmentation contributes to actin cytoskeletal collapse, nucleolysis, and apoptosis of vascular smooth muscle cells exposed to proinflammatory cytokines.
Eur J Cell Biol
77:
294-302,
1998[ISI][Medline].
20.
Goldblum, SE,
Ding X,
and
Campbell-Washington J.
TNF- induces endothelial cell F-actin depolymerization, new actin synthesis, and barrier dysfunction.
Am J Physiol Cell Physiol
264:
C894-C905,
1993
21.
Goldblum, SE,
Hennig B,
Jay M,
Yoneda K,
and
McClain CJ.
Tumor necrosis factor--induced pulmonary vascular endothelial injury.
Infect Immun
57:
1218-1226,
1989[ISI][Medline].
22.
Goldblum, SE,
and
Sun WL.
Tumor necrosis factor- augments pulmonary arterial transendothelial albumin flux in vitro.
Am J Physiol Lung Cell Mol Physiol
258:
L57-L67,
1990
23.
Goldman, G,
Welbourn R,
Kobzik L,
Valeri CR,
Shepro D,
and
Hechtman HB.
Lavage with leukotriene B4 induces lung generation of tumor necrosis factor-alpha that in turn mediates neutrophil diapedesis.
Surgery
113:
297-303,
1993[ISI][Medline].
24.
Haimovitz-Friedman, A,
Cordon-Cardo C,
Bayoumy S,
Garzotto M,
McLoughlin M,
Gallily R,
Edwards CK,
Schuchman EH,
Fuks Z,
and
Kolesnick R.
Lipopolysaccharide induces disseminated endothelial apoptosis requiring ceramide generation.
J Exp Med
186:
1831-1841,
1997
25.
Hall, AK.
Thymosin beta-10 accelerates apoptosis.
Cell Mol Biol Res
41:
167-180,
1995[ISI][Medline].
26.
Hashimoto, S,
Kobayashi A,
Kooguchi K,
Kitamura Y,
Onodera H,
and
Nakajima H.
Upregulation of two death pathways of perforin/granzyme and FasL/Fas in septic acute respiratory distress syndrome.
Am J Respir Crit Care Med
161:
237-243,
2000
27.
Hocking, DC,
Phillips PG,
Ferro TJ,
and
Johnson A.
Mechanisms of pulmonary edema induced by tumor necrosis factor-alpha.
Circ Res
67:
68-77,
1990[Abstract].
28.
Huot, J,
Houle F,
Rousseau S,
Deschesnes RG,
Shah GM,
and
Landry J.
SAPK2/p38-dependent F-actin reorganization regulates early membrane blebbing during stress-induced apoptosis.
J Cell Biol
143:
1361-1373,
1998
29.
Hyers, TM,
Tricomi SM,
Dettenmeier PA,
and
Fowler AA.
Tumor necrosis factor levels in serum and bronchoalveolar lavage fluid of patients with the adult respiratory distress syndrome.
Am Rev Respir Dis
144:
268-271,
1991[ISI][Medline].
30.
Karsan, A,
Yee E,
and
Harlan JM.
Endothelial cell death induced by tumor necrosis factor-alpha is inhibited by the Bcl-2 family member, A1.
J Biol Chem
271:
27201-27204,
1996
31.
Kawai, T,
Matsumoto M,
Takeda K,
Sanjo H,
and
Akira S.
ZIP kinase, a novel serine/threonine kinase which mediates apoptosis.
Mol Cell Biol
18:
1642-1651,
1998
32.
Kayalar, C,
Ord T,
Testa MP,
Zhong LT,
and
Bredesen DE.
Cleavage of actin by interleukin 1-converting enzyme to reverse DNase I inhibition.
Proc Natl Acad Sci USA
93:
2234-2238,
1996
33.
Khimenko, PL,
Moore TM,
Wilson PS,
and
Taylor AE.
Role of calmodulin and myosin light-chain kinase in lung ischemia-reperfusion injury.
Am J Physiol Lung Cell Mol Physiol
271:
L121-L125,
1996
34.
Koga, S,
Morris S,
Ogawa S,
Liao H,
Bilezikian JP,
Chen G,
Thompson WJ,
Ashikaga T,
Brett J,
Stern DM,
and
Pinsky DJ.
TNF modulates endothelial properties by decreasing cAMP.
Am J Physiol Cell Physiol
268:
C1104-C1113,
1995
35.
Koseki, T,
Inohara N,
Chen S,
and
Nunez G.
ARC, an inhibitor of apoptosis expressed in skeletal muscle and heart that interacts selectively with caspases.
Proc Natl Acad Sci USA
95:
5156-5160,
1998
36.
Kothakota, S,
Azuma T,
Reinhard C,
Klippel A,
Tang J,
Chu K,
McGarry TJ,
Kirschner MW,
Koths K,
Kwiatkowski DJ,
and
Williams LT.
Caspase-3-generated fragment of gelsolin: effector of morphological change in apoptosis.
Science
278:
294-298,
1997
37.
Laster, SM,
and
Mackenzie JM, Jr.
Bleb formation and F-actin distribution during mitosis and tumor necrosis factor-induced apoptosis.
Microsc Res Tech
34:
272-280,
1996[ISI][Medline].
38.
Lazar, V,
and
Garcia JG.
A single human myosin light chain kinase gene (MLCK; MYLK).
Genomics
57:
256-267,
1999[ISI][Medline].
39.
Levkau, B,
Herren B,
Koyama H,
Ross R,
and
Raines EW.
Caspase-mediated cleavage of focal adhesion kinase pp125FAK and disassembly of focal adhesions in human endothelial cell apoptosis.
J Exp Med
187:
579-586,
1998
40.
Mashima, T,
Naito M,
Fujita N,
Noguchi K,
and
Tsuruo T.
Identification of actin as a substrate of ICE and an ICE-like protease and involvement of an ICE-like protease but not ICE in VP-16-induced U937 apoptosis.
Biochem Biophys Res Commun
217:
1185-1192,
1995[ISI][Medline].
41.
McDonald, DM.
Endothelial gaps and permeability of venules in rat tracheas exposed to inflammatory stimuli.
Am J Physiol Lung Cell Mol Physiol
266:
L61-L83,
1994
42.
McDonald, DM,
Thurston G,
and
Baluk P.
Endothelial gaps as sites for plasma leakage in inflammation.
Microcirculation
6:
7-22,
1999[ISI][Medline].
43.
Medina, J,
Baud L,
Garcia Escribano C,
Gila JA,
Rodriguez Puyol D,
and
Rodriguez Puyol M.
Tumor necrosis factor-induced contraction of cultured rat mesangial cells: interaction with angiotensin II.
J Lab Clin Med
122:
164-172,
1993[ISI][Medline].
44.
Messam, CA,
and
Pittman RN.
Asynchrony and commitment to die during apoptosis.
Exp Cell Res
238:
389-398,
1998[ISI][Medline].
45.
Miao, JY,
Araki S,
Kaji K,
and
Hayashi H.
Integrin 4 is involved in apoptotic signal transduction in endothelial cells.
Biochem Biophys Res Commun
233:
182-186,
1997[ISI][Medline].
46.
Millar, AB,
Foley NM,
Singer M,
Johnson NM,
Meager A,
and
Rook GA.
Tumour necrosis factor in bronchopulmonary secretions of patients with adult respiratory distress syndrome.
Lancet
2:
712-714,
1989[ISI][Medline].
47.
Mills, JC,
Stone NL,
Erhardt J,
and
Pittman RN.
Apoptotic membrane blebbing is regulated by myosin light chain phosphorylation.
J Cell Biol
140:
627-636,
1998
48.
Morel, NM,
Petruzzo PP,
Hechtman HB,
and
Shepro D.
Inflammatory agonists that increase microvascular permeability in vivo stimulate cultured pulmonary microvessel endothelial cell contraction.
Inflammation
14:
571-583,
1990[ISI][Medline].
49.
Partridge, CA,
Horvath CJ,
Del Vecchio PJ,
Phillips PG,
and
Malik AB.
Influence of extracellular matrix in tumor necrosis factor-induced increase in endothelial permeability.
Am J Physiol Lung Cell Mol Physiol
263:
L627-L633,
1992
50.
Patterson, CE,
Davis HW,
Schaphorst KL,
and
Garcia JG.
Mechanisms of cholera toxin prevention of thrombin- and PMA-induced endothelial cell barrier dysfunction.
Microvasc Res
48:
212-235,
1994[ISI][Medline].
51.
Patterson, CE,
Stasek JE,
Schaphorst KL,
Davis HW,
and
Garcia JG.
Mechanisms of pertussis toxin-induced barrier dysfunction in bovine pulmonary artery endothelial cell monolayers.
Am J Physiol Lung Cell Mol Physiol
268:
L926-L934,
1995
52.
Petrache, I,
Verin AD,
and
Garcia J.
Role of myosin light chain phosphorylation in TNF-alpha induced apoptosis of pulmonary endothelial cells (Abstract).
Am J Respir Crit Care Med
159:
A217,
1999[ISI].
53.
Polunovsky, VA,
Wendt CH,
Ingbar DH,
Peterson MS,
and
Bitterman PB.
Induction of endothelial cell apoptosis by TNF-: modulation by inhibitors of protein synthesis.
Exp Cell Res
214:
584-594,
1994[ISI][Medline].
54.
Robaye, B,
Mosselmans R,
Fiers W,
Dumont JE,
and
Galand P.
Tumor necrosis factor induces apoptosis (programmed cell death) in normal endothelial cells in vitro.
Am J Pathol
138:
447-453,
1991[Abstract].
55.
Saito, H,
Minamiya Y,
Kitamura M,
Saito S,
Enomoto K,
Terada K,
and
Ogawa J.
Endothelial myosin light chain kinase regulates neutrophil migration across human umbilical vein endothelial cell monolayer.
J Immunol
161:
1533-1540,
1998
56.
Slowik, MR,
Min W,
Ardito T,
Karsan A,
Kashgarian M,
and
Pober JS.
Evidence that tumor necrosis factor triggers apoptosis in human endothelial cells by interleukin-1-converting enzyme-like protease-dependent and -independent pathways.
Lab Invest
77:
257-267,
1997[ISI][Medline].
57.
Song, Q,
Wei T,
Lees-Miller S,
Alnemri E,
Watters D,
and
Lavin MF.
Resistance of actin to cleavage during apoptosis.
Proc Natl Acad Sci USA
94:
157-162,
1997
58.
Spyridopoulos, I,
Brogi E,
Kearney M,
Sullivan AB,
Cetrulo C,
Isner JM,
and
Losordo DW.
Vascular endothelial growth factor inhibits endothelial cell apoptosis induced by tumor necrosis factor-: balance between growth and death signals.
J Mol Cell Cardiol
29:
1321-1330,
1997[ISI][Medline]. [Corrigenda. J Mol Cell Cardiol 30: April 1998, p. 897.]
59.
Stelzner, TJ,
Weil JV,
and
O'Brien RF.
Role of cyclic adenosine monophosphate in the induction of endothelial barrier properties.
J Cell Physiol
139:
157-166,
1989[ISI][Medline].
60.
Suarez-Huerta, N,
Lecocq R,
Mosselmans R,
Galand P,
Dumont JE,
and
Robaye B.
Myosin heavy chain degradation during apoptosis in endothelial cells.
Cell Prolif
33:
101-114,
2000[ISI][Medline].
61.
Suarez-Huerta, N,
Mosselmans R,
Dumont JE,
and
Robaye B.
Actin depolymerization and polymerization are required during apoptosis in endothelial cells.
J Cell Physiol
184:
239-245,
2000[ISI][Medline].
62.
Suter, PM,
Suter S,
Girardin E,
Roux-Lombard P,
Grau GE,
and
Dayer JM.
High bronchoalveolar levels of tumor necrosis factor and its inhibitors, interleukin-1, interferon, and elastase, in patients with adult respiratory distress syndrome after trauma, shock, or sepsis.
Am Rev Respir Dis
145:
1016-1022,
1992[ISI][Medline].
63.
Tonnesen, MG.
Neutrophil-endothelial cell interactions: mechanisms of neutrophil adherence to vascular endothelium.
J Invest Dermatol
93:
53S-58S,
1989[Abstract].
64.
Verin, AD,
Lazar V,
Torry RJ,
Labarrere CA,
Patterson CE,
and
Garcia JG.
Expression of a novel high molecular weight myosin light chain kinase in endothelium.
Am J Respir Cell Mol Biol
19:
758-766,
1998
65.
Watson, RW,
Rotstein OD,
Parodo J,
Jimenez M,
Soric I,
Bitar R,
and
Marshall JC.
Impaired apoptotic death signaling in inflammatory lung neutrophils is associated with decreased expression of interleukin-1 converting enzyme family proteases (caspases).
Surgery
122:
163-171,
1997[ISI][Medline].
66.
Waxman, AB,
Einarsson O,
Seres T,
Knickelbein RG,
Warshaw JB,
Johnston R,
Homer RJ,
and
Elias JA.
Targeted lung expression of interleukin-11 enhances murine tolerance of 100% oxygen and diminishes hyperoxia-induced DNA fragmentation.
J Clin Invest
101:
1970-1982,
1998
67.
Wojciak-Stothard, B,
Entwistle A,
Garg R,
and
Ridley AJ.
Regulation of TNF--induced reorganization of the actin cytoskeleton and cell-cell junctions by Rho, Rac, and Cdc42 in human endothelial cells.
J Cell Physiol
176:
150-165,
1998[ISI][Medline].
68.
Wong, RK,
Baldwin AL,
and
Heimark RL.
Cadherin-5 redistribution at sites of TNF-- and IFN-
-induced permeability in mesenteric venules.
Am J Physiol Heart Circ Physiol
276:
H736-H748,
1999
69.
Wright, SC,
Zheng H,
Zhong J,
Torti FM,
and
Larrick JW.
Role of protein phosphorylation in TNF-induced apoptosis: phosphatase inhibitors synergize with TNF to activate DNA fragmentation in normal as well as TNF-resistant U937 variants.
J Cell Biochem
53:
222-233,
1993[ISI][Medline].
70.
Yonemaru, M,
Kasuga I,
Kusumoto H,
Kiyokawa H,
Kuwabara S,
Ichinose Y,
and
Toyama K.
Protein kinase inhibitor attenuates an increase in endothelial monolayer permeability induced by tumour necrosis factor-.
Respirology
2:
63-69,
1997[Medline].