You are on page 1of 10

Neutrophils release extracellular DNA traps in

response to exercise
Thomas Beiter, Annunziata Fragasso, Jens Hudemann, Marius Schild, Jürgen
Steinacker, Frank C. Mooren and Andreas M. Niess
J Appl Physiol 117:325-333, 2014. First published 15 May 2014; doi:10.1152/japplphysiol.00173.2014

You might find this additional info useful...

This article cites 58 articles, 22 of which can be accessed free at:


/content/117/3/325.full.html#ref-list-1

This article has been cited by 1 other HighWire hosted articles


Direct measurement of cell-free DNA from serially collected capillary plasma during
incremental exercise
Sarah Breitbach, Björn Sterzing, Carlos Magallanes, Suzan Tug and Perikles Simon
J Appl Physiol, July 15, 2014; 117 (2): 119-130.
[Abstract] [Full Text] [PDF]

Updated information and services including high resolution figures, can be found at:
/content/117/3/325.full.html

Additional material and information about Journal of Applied Physiology can be found at:

Downloaded from on November 5, 2014


http://www.the-aps.org/publications/jappl

This information is current as of November 5, 2014.

Journal of Applied Physiology publishes original papers that deal with diverse areas of research in applied physiology, especially
those papers emphasizing adaptive and integrative mechanisms. It is published 12 times a year (monthly) by the American
Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2014 by the American Physiological Society.
ISSN: 0363-6143, ESSN: 1522-1563. Visit our website at http://www.the-aps.org/.
J Appl Physiol 117: 325–333, 2014.
First published May 15, 2014; doi:10.1152/japplphysiol.00173.2014.

Neutrophils release extracellular DNA traps in response to exercise


Thomas Beiter,1 Annunziata Fragasso,1 Jens Hudemann,1 Marius Schild,2 Jürgen Steinacker,3
Frank C. Mooren,2 and Andreas M. Niess1
1
Department of Sports Medicine, Medical Clinic, Eberhard-Karls-University of Tuebingen; 2Department of Sports Medicine,
Justus-Liebig University, Giessen; and 3Division of Sports and Rehabilitation Medicine, Ulm University Medical
Center, Germany
Submitted 21 February 2014; accepted in final form 8 May 2014

Beiter T, Fragasso A, Hudemann J, Schild M, Steinacker J, dality in a wide variety of chronic inflammatory conditions (28,
Mooren FC, Niess AM. Neutrophils release extracellular DNA traps 57). However, if we indeed consider exercise as a discrete
in response to exercise. J Appl Physiol 117: 325–333, 2014. First therapeutic modality, we also have to consider that—as with
published May 15, 2014; doi:10.1152/japplphysiol.00173.2014.—In- any medical treatment—prescription of exercise mode, inten-
tense exercise evokes a rapid and transient increase in circulating
cell-free DNA (cf-DNA), a phenomenon that is commonly observed
sity, and duration must be based on a careful evaluation of risks
in a variety of acute and chronic inflammatory conditions. In this and benefits, taking into account possible contraindications and
study, we aimed to shed new light on the release and clearance the patient’s individual risk profile. To develop appropriate
mechanisms of cf-DNA in response to exercise. We hypothesized that exercise intervention guidelines, it is inevitable to reach a
activated neutrophils may primarily contribute to exercise-evoked deeper understanding of how, when, and why physical activity
cf-DNA levels by releasing neutrophil extracellular traps (NETs). interferes with basic mechanisms of our immune system.
Analysis of plasma and/or serum samples from male athletes at rest Depending on intensity and duration, acute physical exercise
and in response to exhaustive treadmill exercise revealed an immedi- constitutes a multiple-stressor situation involving oxidative,

Downloaded from on November 5, 2014


ate and transient increase in cf-DNA that was concomitantly counter- metabolic, mechanical, thermal, and neurohormonal stressors
balanced by an increase in serum DNase activity. Consistently, rapid
to trigger mobilization and activation of leucocytes, acute
release and clearance kinetics for cf-DNA could also be observed in
response to intensive cycling exercise, with no significant differences phase proteins, platelet activation, and alterations of throm-
between endurance-trained (V̇O2max ⬎57 ml·min⫺1·kg⫺1) and healthy botic and fibrinolytic pathways (58). Recently, it has been
(V̇O2max ⬍49 ml·min⫺1·kg⫺1) sedentary individuals. In postexercise shown that strenuous exercise likewise provokes an immediate
blood smear samples, we detected seemingly intact neutrophils dis- and transient increase in plasma circulating cell-free DNA
playing morphological signs of NET release, as indicated by abnormal (cf-DNA) (2, 5, 22). Increased concentrations of cf-DNA have
swollen nuclei and emanating DNA fibers. In support, we observed a for a long time been implicated in a wide range of acute and
striking correlation of postexercise cf-DNA concentrations with chronic inflammatory conditions, including cancer, autoim-
plasma levels of the granule-derived enzyme myeloperoxidase. Our mune diseases, trauma, sepsis, stroke, and myocardial infarc-
study indicates that intense exercise induces liberation of NETs, tion (54). As a consequence, cf-DNA is considered to have
which is sufficiently counterbalanced in healthy individuals by a
diagnostic and prognostic power in malignant as well as in
concomitant rise in serum DNase activity. As aberrant release of
NETs has been linked to diverse disease states, monitoring of cf- nonmalignant diseases (32). Remarkably, despite intensive
DNA/DNase levels or activities in response to standardized exercise research over the course of decades, there still is no consensus
testing could provide a valuable tool to identify people who are at on the origin, release mechanisms, and biological significance
increased risk for cardiac ischemia, thrombosis, autoimmunity, or of cf-DNA. Most commonly, the presence of cf-DNA in the
chronic fatigue. circulation has been ascribed to cell death, either directly by
neutrophil extracellular traps; cell-free DNA; DNase
release from necrotic and apoptotic cells, or indirectly by
liberation from macrophages or scavengers following absorp-
tion of dying cells (54).
AT THE ADVENT OF THE 21st century, the prevalence of chronic A novel fascinating explanation of how DNA can actively be
diseases in developed and developing countries is increasing at released under inflammatory conditions has recently become
an alarming rate (37). Chronic conditions like obesity, diabe- apparent by the discovery of an evolutionary highly conserved
tes, and cardiovascular disease contribute largely to morbidity first-line defense mechanism that allows neutrophils to expel
and mortality all over the globe and place increasing pressure their DNA in response to infectious and endogenous stimuli,
on health care systems (4). Because chronic disorders are thereby forming an antimicrobial meshwork of chromatin and
primarily attributable to lifestyle-related factors, a sizable frac- granule proteins, termed neutrophil extracellular traps (NETs)
tion of this disease burden could substantially be minimized by (8). Accumulating evidence indicates that formation of NETs
nonpharmacological interventions. plays a pivotal role in innate immune responses, allowing
Adopting a regular exercise routine is widely recognized as neutrophils to capture, neutralize, and degrade a variety of
offering protection against obesity, insulin resistance, and ath- invading microorganisms (9). Moreover, NETs have been
erosclerosis (28, 57). Moreover, there is accumulating knowl- shown to constitute a scaffold and stimulus for platelet and red
edge that exercise intervention provides a true treatment mo- blood cell adhesion, fibrin deposition, and thrombus formation,
thereby providing a physical barrier to prevent microbial dis-
semination (12, 25, 40). Aside from infection, NETs have also
Address for reprint requests and other correspondence: T. Beiter, Dept. of
Sports Medicine, Medical Clinic, Eberhard-Karls-Univ. of Tuebingen, Hoppe-
been found to be implicated in sterile inflammation, and,
Seyler-Str. 6, 72076 Tuebingen, Germany (e-mail: thomas.beiter@med.uni- besides their beneficial properties, dysregulated NET forma-
tuebingen.de). tion emerges to profoundly contribute to the pathogenesis of
http://www.jappl.org 8750-7587/14 Copyright © 2014 the American Physiological Society 325
326 NETs in Exercise • Beiter T et al.

diverse autoinflammatory conditions, vascular inflammation, both groups are shown in Table 1. Within 4 wk from the initial
and atherogenesis (19, 41, 48, 53). performance diagnostic, the study participants performed the main
By monitoring the time course of cf-DNA plasma concen- exercise protocol that consisted of 60 min of high intensity cycling
trations in healthy individuals during and after incremental exercise on a cycle ergometer (Ergometrics 800 S, Ergoline, Bitz,
Germany) at a power requiring 80% of the V̇O2max (including 10 min
treadmill running, we recently reported rapid release and clear-
of warm-up cycling at 60% V̇O2max). All exercise sessions were
ance characteristics of cf-DNA in response to exhaustive ex- conducted in the morning (9:00 AM) after an overnight fast and after
ercise (5). Based on this observation, we hypothesized that the receiving a standardized breakfast (8:00 AM). Serum and EDTA
immediate exercise-induced increase in cf-DNA might be in- blood samples were collected at rest (pre), immediately after (post),
dicative of NETs that could be formed within the vasculature, and 3 h (⫹3 h) after the cycling exercise protocol. Routine blood
as has recently been observed in septic blood (12, 48, 59). We variables were determined for each time point by standard methods.
further speculated that release of cf-DNA in response to acute Blood samples were centrifuged at 1,600 g for 10 min, followed by
exercise might be efficiently counterbalanced in healthy indi- 5 min of centrifugation at 16,000 g, and aliquots were stored at ⫺80°C
viduals by an increase in serum DNase 1 activity. To test our until further analysis.
hypothesis, we measured cf-DNA concentrations as well as Quantification of cf-DNA. DNA was extracted from 400 ␮l of
plasma according to the blood and body fluid protocol of the QIAamp
DNase 1 activity in the blood of healthy individuals at rest and Blood Mini Kit (Qiagen, Hilden, Germany) with a final elution
in response to intensive endurance exercise. As regular phys- volume of 100 ␮l. cf-DNA equivalents were quantified by SYBR
ical activity is generally considered to exert beneficial effects green real-time PCR that targeted an 88-bp fragment of the chromo-
on immune functions, we included sedentary as well as well- somal myostatin (MSTN) gene locus, as described previously (5).
trained individuals in our study. In addition, we quantified Briefly, samples were run in triplicates on an iCycler single-color
plasma markers of inflammation, muscle injury, and neutrophil detection system (BioRad, Munich, Germany) using the QuantiFast
activation. We further attempted to visualize exercise-induced SYBR Green PCR Kit (Qiagen). DNA levels were calculated from a
NET formation in the bloodstream by immunocytochemical standard curve generated from serial dilutions of a genomic calibrator
by plotting the threshold cycle value against the logarithm of calibra-

Downloaded from on November 5, 2014


staining of blood smear samples. Our data indicate that intra-
vascular NETs are released during intensive exercise. tor copy numbers. Copies of the target sequence per plasma sample
volume were then calculated as described by Chiu et al. (11), and total
MATERIALS AND METHODS quantities of cf-DNA are given as genome equivalents (GE) per
milliliter of plasma, with one GE being defined as the amount of the
Study design. In the first part of the study, we attempted to provide target sequence contained in a single haploid cell. The lower quanti-
initial evidence for the proposed release and clearance mechanisms of fication and detection limits of the method were reproducibly identi-
cf-DNA. We chose an exhaustive treadmill protocol that has recently fied at 500 GE/ml and 250 GE/ml plasma, respectively.
been shown as suitable for provoking immediate and pronounced Quantification of serum DNase 1 activity. DNase activity in serum
increases in cf-DNA (5). The second part of the study aimed to samples was determined by using Org 590 DNase activity ELISA
confirm these results in a different exercise setting that allowed (Orgentec, Mainz, Germany) according to the manufacturer’s instruc-
assessment of whether the outcome variables would be influenced by tions. This assay measures the intrinsic DNA degradation capacity of
the training status. All experimental procedures were approved by the a serum sample by the turnover of an immobilized DNase substrate
Institutional Review Board of the University of Tuebingen according coated on the chamber surfaces of a microwell plate. Remaining
to the Declaration of Helsinki. substrate is quantified photometrically at 450 nm, and the amount of
Study participants and exercise setups. All subjects were fully color reaction is inversely proportional to the DNase activity in the
informed of the experimental protocols and gave written informed serum sample (activity reduction). To make data more amenable for
consent before the study commenced. Vital signs, anthropometric quantitative comparison, we included a standard curve [5.31 up to
measurements, and physical examinations were conducted to evaluate 277.53 milli-Kunitz units (mKuU)/ml] with defined activities of a
health status. Subjects were excluded if they presented with any acute recombinant human DNase 1 standard provided by the manufacturer.
or chronic disease, major or minor injuries, or use of prescribed Staining of blood smears. Blood droplets (35 ␮l) from EDTA
medication. In the initial study part, six well-trained male athletes whole blood samples were placed on glass slides and gently spread
(median age, 24 yr; range, 20 –33 yr) who were rerecruited from our using the edge of a second glass slide. For histological staining, blood
previous study (5) repeated a graded exercise test until volitional smears were air-dried, fixed in methanol, and stained by standard
exhaustion on a motorized treadmill (Saturn, HP COSMOS, Traun- Giemsa stain (Carl Roth, Karlsruhe, Germany) procedure.
stein, Germany) at a constant incline of 1% with an increment of 2 For immunohistochemical staining of neutrophils, blood smears
km/h every 3 min. Serum and EDTA blood samples were taken before were fixed in 4% paraformaldehyde in PBS for 2 h at room temper-
(pre), immediately after (post), and 30 min (⫹30 min) after the run. ature, rinsed with PBS, permeabilized with 0.5% Triton X-100 PBS
To determine effects of training status, healthy male subjects were for 1 min, and blocked overnight in 2% BSA PBS at 4°C. Incubation
recruited who were either engaged in regular endurance exercise of
more than 5 h/wk or habitually spent less than 2 h/wk with physical
activity. All subjects underwent preliminary baseline screening in- Table 1. Physical characteristics of endurance-trained
cluding lactate diagnostic and maximal bicycle spiroergometry to and untrained participants
determine maximal oxygen uptake (V̇O2max) as defined by the highest
oxygen uptake rate measured during the test, and expressed as V̇O2max, Power Output at 80%
Age, yr BMI, kg/m2 ml 䡠 min⫺1 䡠 kg V̇O2max, Watt
milliliters of oxygen used in one minute per kilogram of body weight
(ml·min⫺1·kg body wt⫺1). The power (in watts) performed at the time ET 27.0 22.7 65.4 250
of 100%, 80%, and 60% V̇O2max was recorded. Depending on their (n ⫽ 6) (26–30) (21–24) (58–67)* (230–310)*
endurance capacity, six sedentary or untrained individuals (UT) and UT 26.5 21.9 39.5 150
six endurance-trained individuals (ET) who met the final inclusion (n ⫽ 6) (19–28) (20–25) (30–48) (50–190)
criteria of V̇O2max ⬍49 ml·min⫺1·kg body wt⫺1 for the UT group and Values are median (range); BMI, body mass index; V̇O2max, maximal
V̇O2max ⬎57 ml·min⫺1·kg body wt⫺1 for the ET group, respectively, aerobic capacity; ET, endurance trained; UT, untrained. *P ⬍ 0.01, signifi-
were included to take part in the study. The anthropometric data for cantly different compared with UT group.

J Appl Physiol • doi:10.1152/japplphysiol.00173.2014 • www.jappl.org


NETs in Exercise • Beiter T et al. 327
with primary rabbit anti-human neutrophil elastase antibodies 15.2-fold, from 45.1 (29.4 –79.4) mKuU/ml at rest to 310.9
(ab68672, 1:200; Abcam, Cambridge, United Kingdom) or the corre- (187.6 –594.3) mKuU/ml (P ⫽ 0.005) postexercise (Fig. 1B).
sponding negative control antibodies (Ab 46540, Abcam) was per- Increased serum enzyme activity was of a transient nature and
formed for 1 h at room temperature in 2% BSA PBS, followed by
decreased to preexercise levels at 30 min after cessation of
secondary staining with Cy3-conjugated donkey anti-rabbit IgG (711-
166-152, 1:400, Dianova, Hamburg, Germany) for 1 h at room exercise (median 47.7 mKuU/ml; range 33.0 –194.2).
temperature. DNA was labeled with 4,6-diamidino-2-phenylindole Postexercise blood smears reveal NET-like structures. Con-
(1.25 ␮g/ml; Invitrogen, Life Technologies, Darmstadt, Germany). sidering the rapid appearance of excess cf-DNA in the circu-
Specimens were mounted with Vectashield (Vector Laboratories, lation of exercising subjects, we analyzed Giemsa-stained
Peterborough, United Kingdom), and neutrophils were visualized by blood smears from pre- and postexercise blood samples to
immunofluorescence microscopy (Axiovert, Zeiss, Goettingen, Ger- screen for suspicious signs of damaged or dying blood cells. In
many). peripheral blood smear slides prepared immediately after the
Quantification of plasma markers. Quantification of 89 inflamma-
tion-associated proteins in plasma samples was performed at Rules
exhaustive treadmill run, we observed the scattered but striking
Based Medicine (Myriad RBM, Austin, TX) using their human MAP appearance of seemingly disintegrated cells with a decon-
v1.6 panel of Luminex-based multiplex assays as described by the densed chromatin structure (Fig. 2A). Similar structures are
vendor. Analytes containing values below the lower detection limit apparent in blood smears from patients with malaria (3), and
were excluded from analysis. have recently been documented in blood smears from stored
Plasma volume changes. Study participants were instructed to red blood cell units (24). Here, these cells were recognized as
drink water at two defined time points during the cycling exercise circulating neutrophils that are releasing extracellular DNA
protocol. To confidently exclude biased measures due to plasma
webs, termed NETs. To obtain proof of principle, we used
volume shift, changes in plasma concentrations were calculated from
hemoglobin and hematocrit values and concentrations of postexercise immunocytochemistry to visualize intravascular neutrophils by
variables were corrected according to Kraemer and Brown (35). For neutrophil elastase staining and to clearly characterize NET-
all analyzed postexercise samples, calculated losses in plasma volume like structures in postexercise blood smears. As shown in Fig.

Downloaded from on November 5, 2014


were below 6% (range: 1.5% to 5.4%). 2, suspicious NET-like structures emerged as seemingly intact
Statistical analysis. Statistical analysis was performed using JMP neutrophils with abnormal swollen nuclei and dispersed nu-
(version 10; SAS Institute, Cary, NC) and SPSS statistical software clear staining patterns that were surrounded by a delicate
(version 15; SPSS, Chicago, IL). Differences within groups over time network of emanating DNA (Fig. 2B). Control stainings of
were analyzed by Friedman’s two-way nonparametric ANOVA, fol-
lowed by Wilcoxon’s matched-pairs signed-rank test. The Mann-
isolated neutrophils treated with phorbol myristate acetate
Whitney U-test was used to compare between group variables. Spear- (PMA; data not shown) exemplified that the observed pheno-
man rank correlation coefficients (CCs) were calculated with Bonfer- type is clearly distinct from the commonly described lytic
roni-adjusted significance levels to estimate the correlation between mechanism of NET release that culminates in the rupture of the
variables. A value of P ⬍ 0.05 was regarded as statistically signifi- cellular membrane and the concomitant expulsion of filamen-
cant. Data are presented as median (range). tous chromatin structures (8), but could rather be attributed to
the recently discovered rapid mechanism of vesicular-mediated
RESULTS
NET release (48).
Release of cf-DNA in response to acute exercise is counter- Trained and untrained healthy individuals do not differ in
balanced by serum DNase 1. An immediate, pronounced, and their cf-DNA/DNase responses to exercise. Because aberrant
transient increase in cf-DNA levels could be observed in six release of cf-DNA/NETs has been implicated in diverse
healthy young athletes in response to an acute bout of exhaus- chronic inflammatory conditions, we wanted to address
tive treadmill exercise, rising by 14.9-fold from median base- whether sedentary (untrained, UT) and habitually exercising
line values of 1,645 (1,262– 4,527) GE/ml at rest to 25,511 (endurance-trained, ET) individuals display distinct cf-DNA/
(10,673–54,383) GE/ml (P ⫽ 0.005) at the end of the run, and DNase responses to acute exercise. To achieve comparable
declining to 4.1-fold baseline concentrations after 30 min of workload and duration for both groups, we chose a 1-h high-
recovery (median 7,718 GE/ml; range 4,156 –16,730; P ⫽ intensity cycling protocol adjusted to the individual V̇O2max
0.008; Fig. 1A). Rapid clearance kinetics of cf-DNA could be level. Both groups did not differ in their baseline cf-DNA
attributed to an immediate rise in serum DNase 1 activity by levels, with a median plasma concentration of 2,078 (638 –

A B
60000 ** ** 700 ** **
Plasma DNase activity (mKuU/ml)

50000 600
Plasma cf-DNA (GE/ml)

Fig. 1. Values of cell-free DNA (cf-DNA)


500 plasma concentrations (A) and serum DNase 1
40000 activity (B) in the blood of well-trained individ-
400 uals (n ⫽ 6), determined at rest (pre), in re-
30000 sponse to exhaustive treadmill exercise (post),
300
and after 30 min of recovery (⫹30 min). Bar,
20000 median of the group. **P ⱕ 0.01, significant
200
difference between sampling points.
10000 100

0 0
pre post +30 min pre post +30 min

J Appl Physiol • doi:10.1152/japplphysiol.00173.2014 • www.jappl.org


328 NETs in Exercise • Beiter T et al.

4,197) GE/ml in the ET group and 1,582 (1,240 –3,465) GE/ml exercise (Spearman’s CC ⫽ 0.699, P ⫽ 0.011; Fig. 3E). Of
in the UT group (Fig. 3A). Likewise, there was no difference in note, no significant association could be observed with the
postexercise levels as both groups showed an equally pro- muscle damage marker myoglobin (Spearman’s CC ⫽ ⫺0.028,
nounced increase in cf-DNA, rising significantly by 5.9-fold in P ⫽ 0.931; Fig. 3F). Pronounced liberation of myoglobin
the ET group to 9,425 (5,765–17,549) GE/ml (P ⫽ 0.004) and became apparent at 3 h after cessation of exercise (Fig. 3D),
by 5.8-fold in the UT group to 10,934 (7,525–18,774) GE/ml with a significant correlation to FABP plasma levels (Spear-
(P ⫽ 0.004), respectively. Three hours after cessation of man’s CC ⫽ 0.937, P ⬍ 0.0001), another early cytoplasmic
exercise, cf-DNA plasma concentrations in both the ET group muscle damage marker. Because MPO is particularly abundant
(median 2,948 GE/ml; range 1,515– 8,489) as well as in the UT in the primary granules of neutrophils, correlation of cf-DNA
group (median 2,131 GE/ml; range 1,376 –3,673) had returned and MPO levels in postexercise samples further supports the
to preexercise levels. The same pattern was visible for serum idea that netting neutrophils might substantially contribute to
DNase 1 activity (Fig. 3B). Significant enhanced activity by the immediate cf-DNA release in response to exercise.
8.1-fold and 7.1-fold, respectively, could be observed in re-
sponse to exercise for both the ET group (P ⫽ 0.004) as well
DISCUSSION
as the UT group (P ⫽ 0.002), with median baseline values of
19.3 (13.5–23.6) mKuU/ml and 19.2 (17.4 –25.9) mKuU/ml, Increased concentrations of cf-DNA are frequently observed
respectively, and median postexercise values of 143.6 (122.9 – in acute and chronic inflammatory conditions (54). While the
296.3) mKuU/ml and 133.3 (56.8 –198.1) mKuU/ml, respec- prognostic and diagnostic value of this phenomenon has been
tively. A decline to near baseline activity was consistently extensively attributed in diverse malignancies as well as in
observed in both groups after 3 h of recovery, with median infectious and autoinflammatory diseases (32), the biological
serum activities of 23.0 (14.1–97.2) mKuU/ml and 30.3 (25.9 – significance of nonpathological release of cf-DNA in the
130.1) mKuU/ml respectively.
course of physical exercise remains enigmatic. It has initially

Downloaded from on November 5, 2014


Release of cf-DNA in response to exercise is associated with
been assumed that leakage of nuclear content from traumatized
increased plasma myeloperoxidase levels. To further charac-
terize the inflammatory milieu of exercise-induced release of muscle fibers may primarily account for exercise-induced cf-
cf-DNA, we determined plasma concentrations of 89 inflam- DNA levels, thus simply representing a bystander marker of
mation-associated proteins using a multiplex quantification muscle damage (2). This view has recently been challenged by
approach. The analytes that could be found significantly in- thorough time-course analyses, showing clearly distinct release
creased in the circulation of either group at any of the sampling kinetics of cf-DNA and cytosolic markers of muscle damage
points following exercise were the interleukins IL-6, IL-8, during and following strenuous endurance exercise (5, 23).
IL-10, and IL-16; growth hormone; the secretable matrix Here, we provide evidence that release of cf-DNA in response
metalloproteinases MMP2, MMP3, and MMP9; the myeloid to exercise rather reflects an active effector mechanism of
cell-specific enzyme myeloperoxidase (MPO; Fig. 3E); as well innate immunity, namely the formation of NETs by peripheral
as myoglobin (Fig. 3D) and the heart-type fatty acid-binding blood neutrophils. Our results show that 1) exercise-induced
protein (FABP), both representing cytoplasmic muscle constit- cf-DNA is efficiently counterbalanced in healthy individuals
uents (Table 2). Correlation analysis revealed that MPO was by a concomitant increase in serum DNase 1 activity; 2) the
the only analyte with postexercise plasma levels significantly immediate appearance of cf-DNA in the circulation clearly
correlated with the amount of cf-DNA released in response to precedes and is not correlated with the early muscle damage

Fig. 2. Representative images of neutrophil extra-


cellular trap (NET)-like structures in the postexer-
cise blood of a well-trained subject. Blood smears
were prepared immediately after exhaustive tread- A B
mill exercise and Giemsa stained for cytological NE NE NE
examination (A). Intact neutrophils are clearly de-
lineated by their lobular and segmented nuclei (A,
overview image and top left). NET-like structures
appear as seemingly disintegrated cells with a
decondensed chromatin structure and fibrous pro-
trusions of DNA (A, overview image and top DAPI DAPI DAPI
right). Blood smears were further analyzed by
immunocytochemistry for neutrophil elastase
(NE; B). Bottom row panels show merged immu-
nofluorescence images of representative neutro-
phils in postexercise blood smears. NE is shown in
red (top row). DNA was labeled with 4,6-di-
amidino-2-phenylindole (DAPI) and is shown in Merge Merge Merge
blue (middle row). Intact neutrophils were identi-
fied as NE-positive cells with typical segmented
nuclei (B, left). Netting neutrophils were charac-
terized as NE-positive cells with seemingly intact
membrane structure and disintegrated nuclei (B,
middle and right columns), and NET-like struc-
tures were identified as delicate networks of ex-
tracellular DNA. Bar, 10 ␮m.

J Appl Physiol • doi:10.1152/japplphysiol.00173.2014 • www.jappl.org


NETs in Exercise • Beiter T et al. 329

A ** ** B **
20000 ** * ** *
350

Plasma DNase activity (mKuU/ml)


300
Plasma cf-DNA (GE/ml)

15000
250

200
10000
150

5000 100

50

0 0
ET UT ET UT ET UT ET UT ET UT ET UT
pre post +3h pre post +3h Fig. 3. Effect of acute exercise and training
status on plasma levels of cf-DNA (A), serum

C ** D ** DNase activity (B), plasma concentrations of


myeloperoxidase (MPO; C), and myoglobin
300 * 1000 ** (D). Values were retrieved from blood samples
of endurance-trained (ET; n ⫽ 6; closed circles;
Plasma myoglobin (ng/ml)

250 800 V̇O2max ⬎57 ml·min⫺1·kg body wt⫺1) and sed-


Plasma MPO (ng/ml)

entary (untrained, UT; open circles; n ⫽ 6;


200 V̇O2max ⬍49 ml·min⫺1·kg body wt⫺1) individ-
600
uals at rest (pre), after 60 min of intense cycling

Downloaded from on November 5, 2014


150 exercise (post), and after 3 h of recovery (⫹3
400 h). Bar, median of the group. A significant
100 positive association between plasma cf-DNA
200 levels and MPO concentrations could be de-
50 tected in postexercise samples (E), while no
significant correlation could be seen between
0 0 cf-DNA levels and the muscle damage marker
ET UT ET UT ET UT ET UT ET UT ET UT myoglobin (F). mKuU, milli-Kunitz units; GE,
pre post +3h pre post +3h genome equivalents. *P ⬍ 0.05, significant dif-
ference within group. **P ⬍ 0.01, significant
difference within group.
E F
25000 25000
Spearman‘s rho = 0.699, P = 0.011 Spearman‘s rho = -0.028, P = 0.931
cf-DNA (GE/ml plasma)
cf-DNA (GE/ml plasma)

20000 20000

15000 15000

10000 10000

5000 5000

0 0
100 150 200 250 300 20 30 40 50 60 70 80 90
MPO (ng/ml plasma) Myoglobin (ng/ml plasma)

markers myoglobin and FABP; 3) postexercise concentra- erated by exocytosis (43, 52). As indicated by the observed
tions of cf-DNA are significantly correlated with circulating association between plasma levels of cf-DNA and MPO, it
levels of the neutrophil granule enzyme MPO; and 4) has become evident that active release of MPO by neutro-
neutrophils with delobulated nuclei and emanating DNA phils is primarily accomplished via colocalization with the
fiber structures are apparent in postexercise blood smears, chromatin backbone of NETs (55). Consistently, levels of
indicating that intravascular NETs are released in response MPO have been shown to strongly correlate with cf-DNA in
to intense exercise. the blood of patients with thrombotic microangiopathies
In line with our results, degranulation of neutrophils, as (26), and in patients with deep vein thrombosis (17). More-
predicted by an immediate increase of MPO in the circula- over, by use of capture ELISA protocols, true MPO-DNA
tion, has commonly been reported in response to intense complexes have recently been detected in the circulation of
physical exercise (46, 47). Remarkably, the hemoprotein patients with small vessel vasculitis (SVV) (34) and in
MPO is contained within the neutrophil primary granules, individuals with suspected coronary artery disease (6).
which are destined to fuse with the forming or formed NETs were originally described as an innate defense mech-
phagosome after microbial engulfment, but are barely lib- anism to extracellularly capture and potentially kill invading

J Appl Physiol • doi:10.1152/japplphysiol.00173.2014 • www.jappl.org


330 NETs in Exercise • Beiter T et al.

Table 2. Plasma concentrations of inflammation-associated blood markers at rest, immediately after, and 3 h after 60 min
of intense cycling exercise in endurance-trained and untrained individuals
Pre Post ⫹3 h

ET UT ET UT ET UT

FABP, ng/ml 2.4 1.6 4.1 2.5 4.1 11.1


(1.4–3.4) (1.4–2.4) (2.8–7.3) (1.8–3.4) (3.7–6.9)*,† (2.0–49.4)‡
GH, ng/ml 0.3 0.2 18.8 31.0 0.5 0.2
(0.1–0.7) (0.1–0.3) (6.9–99.0)† (8.4–56.8)† (0.2–1.0) (0.1–0.6)
IL-6, pg/ml 1.1 1.1 4.8 3.7 1.5 2.3
(0.1–2.4) (0.1–5.8) (3.6–11.0)† (1.8–5.7) (1.1–2.6) (1.1–4.0)
IL-8, pg/ml 3.5 4.6 7.3 5.5 5.2 4.1
(2.7–6.9) (3.2–5.9) (5.6–9.9)† (2.7–6.5) (4.9–6.2) (0.1–7.1)
IL-10, pg/ml 3.7 4.2 10.6 5.1 5.0 3.8
(2.8–4.6) (1.9–5.6) (5.3–18.9)‡ (2.7–8.9) (3.9–5.7) (2.1–6.1)
IL-16, pg/ml 190 203 359 291 263 278
(147–306) (151–266) (210–589)‡ (217–435) (185–439) (209–415)
Lactate, mmol/l 1.1 0.9 5.3 5.5
(0.7–1.4) (0.7–1.1) (4.2–8.6)‡ (2.3–7.2)‡
MMP2, ng/ml 2,240 2,070 2,585 2,010 2,325 2,056
(1,840–2,510) (1,620–2,650) (2,324–3,401)† (1,530–2,552) (1,982–2,779) (1,482–2,518)
MMP3, ng/ml 12.5 12.5 22.8 14.8 12.6 9.4
(11.0–17.0) (6.5–17.0) (16.9–27.5)† (9.2–23.6) (10.1–17.33) (5.5–19.1)
MMP9, ng/ml 47 45 80 100 92 140
(19–54) (26–78) (69–101) (55–195)† (45–150)† (44–250)†
MPO, ng/ml 105 122 192 190 118 168

Downloaded from on November 5, 2014


(88–156) (105–148) (137–232)† (157–274)‡ (74–204) (119–200)
Myoglobin, ng/ml 34 31 60 47 110 327
(28–72) (27–51) (55–77) (26–81) (73–203)*,‡ (50–914)‡
Values are median (range); n ⫽ 6. Pre, at rest; Post, immediately after; ⫹3h, after 60 min of intense cycling exercise; ET, endurance-trained; UT, untrained;
FABP, fatty acid-binding protein in heart; GH, growth hormone; IL, interleukin; MMP, matrix metalloproteinase; MPO, myeloperoxidase. *P ⬍ 0.05, significant
difference compared to UT group at indicated time point. †P ⬍ 0.05, significant difference within group compared to preexercise level. ‡P ⬍ 0.01, significant
difference within group compared to preexercise level.

pathogens (8, 9). In their seminal study, Brinkmann et al. (8) non-self and altered-self, and has therefore been termed im-
reported that, on stimulation with PMA, neutrophils undergo a munothrombosis (21). NETs have been shown to constitute a
sequel of cytoplasmic and nuclear changes that culminate in scaffold for platelet and red blood cell adhesion and to con-
the rupture of the cellular membrane to release web-like centrate effector proteins involved in thrombus formation (7,
structures of DNA coated with nuclear and granule proteins 25, 56). Moreover, the chromatin backbone of NETs provides
(including MPO). This lytic process of NET release, termed a contact surface to initiate the intrinsic coagulation cascade
NETosis, occurs slowly over 2– 4 h, is dependent on formation (25, 44), and diverse NET components have been shown to
of reactive oxygen species, and is clearly distinct from classical antagonize with natural coagulation inhibitors (1, 39). It is
forms of cell death like apoptosis or necrosis. However, despite noteworthy that strenuous or exhaustive exercise is known to
extensive research efforts in recent years, the molecular path- elicit a transient hypercoagulable state characterized by platelet
ways and the biological significance of this phenomenon are activation as well as selective stimulation of secondary hemo-
just beginning to emerge. It appears that— dependent on stim- stasis via the intrinsic coagulation pathway (20, 36). The
ulus, location, milieu, and activation state—several distinct but physiological implications and mechanistic basis of this spe-
partially overlapping signaling pathways exist that culminate in cific hemostatic response to exercise have been the subject of
the formation of NETs for varying purposes (30, 45). Recently, long-standing controversy, but could indeed be conclusively
Paul Kubes and colleagues (49, 59) uncovered an alternative, described as potentially dependent on the exercise-induced
nonlytic and very rapid process of NET formation that does not formation of intravascular NETs.
require cell death, thereby enabling intravascular neutrophils to Although there are clearly many facets of NETs that require
expel their decondensed nuclear contents without losing their further exploration, it is quite undisputed that appropriate
chemotactic and phagocytic abilities. The described phenotype functioning of NETs is an important facet of effective host
intriguingly resembles that of the seemingly intact NET-form- defense. On the other hand, aberrant or unresolved release of
ing neutrophils we observed in the blood smear specimens of NETs can correlate with disease severity, imposes an increased
postexercise samples. risk of thrombotic events, may contribute to vascular injury
There is increasing evidence that release of chromatin serves and tissue damage, and has been implicated in the etiology of
as an ancient and highly conserved strategy of host defense (9). autoimmunity (9, 30). As complexes of self-DNA and neutro-
It also appears that NETs evolved to interact at different stages phil-derived granule proteins provide a source of autoantigens
of innate immune responses (30). In the vasculature, NETs and have been identified as potent activators of dendritic cells,
recently emerged as being crucially important to accomplish NETs just recently emerged as key players in atherogenesis
bidirectional crosstalk between innate immunity and blood (18, 19) and in the development of diverse autoimmune dis-
coagulation, a task that seems inevitable to protect hosts from eases including systemic lupus erythematosus (SLE) (27),

J Appl Physiol • doi:10.1152/japplphysiol.00173.2014 • www.jappl.org


NETs in Exercise • Beiter T et al. 331
SVV (51), rheumatoid arthritis (RA) (33), inflammatory skin it represents a fundamental layer of immune response to
disease (29), and type I diabetes (16). Moreover, aberrant exercise.
release of NETs has been linked to cancer-associated throm-
bosis, tumor growth, and metastasis (13–15). At first sight, it GRANTS
might appear paradoxical that exercise should trigger an im- Part of this study was supported by the Bundesinstitut für Sportwissen-
mune mechanism that seems crucially involved in many schaft, 081902/09-13.
chronic diseases and conditions that regular exercise is meant
to protect one from. However, one might speculate that, by DISCLOSURES
allowing “safe” release of NETs in a noninflammatory or No conflicts of interest, financial or otherwise, are declared by the author(s).
tolerogenic environment, exercise-induced formation of intra-
vascular NETs might provide an essential and immanent strat- AUTHOR CONTRIBUTIONS
egy to establish self-tolerance to neutrophil-derived peptides Author contributions: T.B., J.M.S., F.C.M., and A.M.N. conception and
and self-DNA, thus contributing significantly to the health design of research; T.B., A.F., and J.H. performed experiments; T.B. and M.S.
benefits of physical activity. analyzed data; T.B., A.F., and A.M.N. interpreted results of experiments; T.B.
Irrespective of its functional role(s), timely removal of prepared figures; T.B. and A.M.N. edited and revised manuscript; T.B., A.F.,
J.H., M.S., J.M.S., F.C.M., and A.M.N. approved final version of manuscript.
exercise-induced NETs should be of indispensable importance
to preserve tissue homeostasis, to avoid thrombotic complica-
REFERENCES
tions, and to prevent aberrant presentation of self-antigens (10,
31, 38, 42). Here, we demonstrate that the rapid resolution of 1. Ammollo CT, Semeraro F, Xu J, Esmon NL, Esmon CT. Extracellular
exercise-induced cf-DNA could be attributed to a concomitant histones increase plasma thrombin generation by impairing thrombomodu-
lin-dependent protein C activation. J Thromb Haemost 9: 1795–1803,
and transient rise in serum DNase 1 activity. DNase 1 is the 2011.
most important waste-management nuclease in serum with 2. Atamaniuk J, Vidotto C, Tschan H, Bachl N, Stuhlmeier KM, Muller

Downloaded from on November 5, 2014


responsibility for degrading extracellular DNA. Consistently, MM. Increased concentrations of cell-free plasma DNA after exhaustive
DNase 1 has been depicted as the primary degrader of NETs, exercise. Clin Chem 50: 1668 –1670, 2004.
3. Baker VS, Imade GE, Molta NB, Tawde P, Pam SD, Obadofin MO,
and impairment or deficiency in DNase 1 activity has been Sagay SA, Egah DZ, Iya D, Afolabi BB, Baker M, Ford K, Ford R,
directly linked to the development of autoimmune diseases (31, Roux KH, Keller TC III. Cytokine-associated neutrophil extracellular
38). Remarkably, an increased seroprevalence of antinuclear traps and antinuclear antibodies in Plasmodium falciparum infected chil-
antibodies, a hallmark of SLE and RA, has also been reported dren under six years of age. Malar J 7: 41, 2008.
4. Bastien M, Poirier P, Lemieux I, Despres JP. Overview of epidemiol-
in athletes suffering from chronic fatigue (50). Thus, it seems ogy and contribution of obesity to cardiovascular disease. Prog Cardio-
reasonable to assume that aberrant or unresolved release of vasc Dis 56: 369 –381, 2014.
cf-DNA/NETs might profoundly contribute to unanticipated 5. Beiter T, Fragasso A, Hudemann J, Niess AM, Simon P. Short-term
adverse outcomes of acute or prolonged exercise in susceptible treadmill running as a model for studying cell-free DNA kinetics in vivo.
Clin Chem 57: 633–636, 2011.
individuals, evoked, for example, by excessive or unaccus- 6. Borissoff JI, Joosen IA, Versteylen MO, Brill A, Fuchs TA, Savchenko
tomed training loads, high stress levels, or insufficient rest AS, Gallant M, Martinod K, Ten CH, Hofstra L, Crijns HJ, Wagner
periods. DD, Kietselaer BL. Elevated levels of circulating DNA and chromatin are
Considering the overall health-promoting effect of regular independently associated with severe coronary atherosclerosis and a pro-
exercise, we wondered whether sedentary and physically active thrombotic state. Arterioscler Thromb Vasc Biol 33: 2032–2040, 2013.
7. Brill A, Fuchs TA, Savchenko AS, Thomas GM, Martinod K, De
individuals in general display distinct cf-DNA/DNase re- Meyer SF, Bhandari AA, Wagner DD. Neutrophil extracellular traps
sponses to acute exercise. Our results indicate that the individ- promote deep vein thrombosis in mice. J Thromb Haemost 10: 136 –144,
ual response to exercise is not directly interconnected to the 2012.
training status, at least not in healthy young individuals— 8. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y,
Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps
which of course does not rule out that exercise intervention kill bacteria. Science 303: 1532–1535, 2004.
might affect these responses in susceptible individuals. Like- 9. Brinkmann V, Zychlinsky A. Neutrophil extracellular traps: is immunity
wise, it will be important to determine the responsiveness in the second function of chromatin? J Cell Biol 198: 773–783, 2012.
individuals with known risk factors for lifestyle-related dis- 10. Caudrillier A, Kessenbrock K, Gilliss BM, Nguyen JX, Marques MB,
Monestier M, Toy P, Werb Z, Looney MR. Platelets induce neutrophil
eases. Remarkably, patients with severe coronary atheroscle- extracellular traps in transfusion-related acute lung injury. J Clin Invest
rosis or calcified coronary arteries have recently been shown to 122: 2661–2671, 2012.
display increased plasma levels of cf-DNA, and quantification 11. Chiu RW, Chan LY, Lam NY, Tsui NB, Ng EK, Rainer TH, Lo YM.
of circulating MPO-DNA complexes has been demonstrated of Quantitative analysis of circulating mitochondrial DNA in plasma. Clin
Chem 49: 719 –726, 2003.
predictive value for future cardiovascular events (6). In this 12. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM,
way, monitoring of cf-DNA/DNase levels or activities in Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, Keys EM, Len-
response to a standardized exercise testing could provide a Vercoe E, Devinney R, Doig CJ, Green FH, Kubes P. Platelet TLR4
valuable tool to identify people who are at increased risk of activates neutrophil extracellular traps to ensnare bacteria in septic blood.
Nat Med 13: 463–469, 2007.
developing cardiac ischemia, thrombosis, autoimmunity, air- 13. Cools-Lartigue J, Spicer J, McDonald B, Gowing S, Chow S, Giannias
way hyperresponsiveness, or chronic fatigue. B, Bourdeau F, Kubes P, Ferri L. Neutrophil extracellular traps seques-
In summary, we provide descriptive evidence of exercise- ter circulating tumor cells and promote metastasis. J Clin Invest 123:
induced release of NETs. This observation might open a new 3446 –3458, 2013.
14. Demers M, Krause DS, Schatzberg D, Martinod K, Voorhees JR,
avenue to improve our understanding of exercise immunology. Fuchs TA, Scadden DT, Wagner DD. Cancers predispose neutrophils to
Future work will show whether this phenomenon is rather a release extracellular DNA traps that contribute to cancer-associated throm-
reminiscence of our ancient fight-or-flight response or whether bosis. Proc Natl Acad Sci USA 109: 13076 –13081, 2012.

J Appl Physiol • doi:10.1152/japplphysiol.00173.2014 • www.jappl.org


332 NETs in Exercise • Beiter T et al.

15. Demers M, Wagner DD. Neutrophil extracellular traps: a new link to 37. Lopez AD, Mathers CD, Ezzati M, Jamison DT, Murray CJ. Global
cancer-associated thrombosis and potential implications for tumor pro- and regional burden of disease and risk factors, 2001: systematic analysis
gression. Oncoimmunology 2: e22946, 2013. of population health data. Lancet 367: 1747–1757, 2006.
16. Diana J, Simoni Y, Furio L, Beaudoin L, Agerberth B, Barrat F, 38. Malickova K, Duricova D, Bortlik M, Hruskova Z, Svobodova B,
Lehuen A. Crosstalk between neutrophils, B-1a cells and plasmacytoid Machkova N, Komarek V, Fucikova T, Janatkova I, Zima T, Lukas
dendritic cells initiates autoimmune diabetes. Nat Med 19: 65–73, 2013. M. Impaired deoxyribonuclease I activity in patients with inflammatory
17. Diaz JA, Fuchs TA, Jackson TO, Hovinga JA, Lammle B, Henke PK, bowel diseases. Autoimmune Dis 2011: 945861, 2011.
Myers DD Jr, Wagner DD, Wakefield TW. Plasma DNA is elevated in 39. Massberg S, Grahl L, von Bruehl ML, Manukyan D, Pfeiler S,
patients with deep vein thrombosis. J Vasc Surg Venous Lymphat Disord Goosmann C, Brinkmann V, Lorenz M, Bidzhekov K, Khandagale
1: 2013. AB, Konrad I, Kennerknecht E, Reges K, Holdenrieder S, Braun S,
18. Doring Y, Drechsler M, Wantha S, Kemmerich K, Lievens D, Vijayan Reinhardt C, Spannagl M, Preissner KT, Engelmann B. Reciprocal
S, Gallo RL, Weber C, Soehnlein O. Lack of neutrophil-derived coupling of coagulation and innate immunity via neutrophil serine pro-
CRAMP reduces atherosclerosis in mice. Circ Res 110: 1052–1056, 2012. teases. Nat Med 16: 887–896, 2010.
19. Doring Y, Manthey HD, Drechsler M, Lievens D, Megens RT, Soehn- 40. McDonald B, Urrutia R, Yipp BG, Jenne CN, Kubes P. Intravascular
lein O, Busch M, Manca M, Koenen RR, Pelisek J, Daemen MJ, neutrophil extracellular traps capture bacteria from the bloodstream during
Lutgens E, Zenke M, Binder CJ, Weber C, Zernecke A. Auto-antigenic sepsis. Cell Host Microbe 12: 324 –333, 2012.
protein-DNA complexes stimulate plasmacytoid dendritic cells to promote 41. Megens RT, Vijayan S, Lievens D, Doring Y, van Zandvoort MA,
atherosclerosis. Circulation 125: 1673–1683, 2012. Grommes J, Weber C, Soehnlein O. Presence of luminal neutrophil
20. El-Sayed MS. Exercise and training effects on platelets in health and extracellular traps in atherosclerosis. Thromb Haemost 107: 597–598,
disease. Platelets 13: 261–266, 2002. 2012.
21. Engelmann B, Massberg S. Thrombosis as an intravascular effector of 42. Meng W, Paunel-Gorgulu A, Flohe S, Witte I, Schadel-Hopfner M,
innate immunity. Nat Rev Immunol 13: 34 –45, 2013. Windolf J, Logters TT. Deoxyribonuclease is a potential counter regu-
22. Fatouros IG, Destouni A, Margonis K, Jamurtas AZ, Vrettou C, lator of aberrant neutrophil extracellular traps formation after major
Kouretas D, Mastorakos G, Mitrakou A, Taxildaris K, Kanavakis E, trauma. Mediators Inflamm 2012: 149560, 2012.
Papassotiriou I. Cell-free plasma DNA as a novel marker of aseptic 43. Nordenfelt P, Tapper H. Phagosome dynamics during phagocytosis by
inflammation severity related to exercise overtraining. Clin Chem 52: neutrophils. J Leukoc Biol 90: 271–284, 2011.
1820 –1824, 2006. 44. Oehmcke S, Morgelin M, Herwald H. Activation of the human contact

Downloaded from on November 5, 2014


23. Fatouros IG, Jamurtas AZ, Nikolaidis MG, Destouni A, Michailidis Y, system on neutrophil extracellular traps. J Innate Immun 1: 225–230,
Vrettou C, Douroudos II, Avloniti A, Chatzinikolaou A, Taxildaris K, 2009.
Kanavakis E, Papassotiriou I, Kouretas D. Time of sampling is crucial 45. Parker H, Winterbourn CC. Reactive oxidants and myeloperoxidase and
for measurement of cell-free plasma DNA following acute aseptic inflam- their involvement in neutrophil extracellular traps. Front Immunol 3: 424,
mation induced by exercise. Clin Biochem 43: 1368 –1370, 2010. 2012.
46. Peake J, Wilson G, Hordern M, Suzuki K, Yamaya K, Nosaka K,
24. Fuchs TA, Alvarez JJ, Martinod K, Bhandari AA, Kaufman RM,
Mackinnon L, Coombes JS. Changes in neutrophil surface receptor
Wagner DD. Neutrophils release extracellular DNA traps during storage
expression, degranulation, and respiratory burst activity after moderate-
of red blood cell units. Transfusion 53: 3210 –3216, 2013.
and high-intensity exercise. J Appl Physiol 97: 612–618, 2004.
25. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M,
47. Peake JM, Suzuki K, Wilson G, Hordern M, Nosaka K, Mackinnon
Myers DD Jr, Wrobleski SK, Wakefield TW, Hartwig JH, Wagner
L, Coombes JS. Exercise-induced muscle damage, plasma cytokines,
DD. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci
and markers of neutrophil activation. Med Sci Sports Exerc 37: 737–
USA 107: 15880 –15885, 2010.
745, 2005.
26. Fuchs TA, Kremer Hovinga JA, Schatzberg D, Wagner DD, Lammle
48. Phillipson M, Kubes P. The neutrophil in vascular inflammation. Nat
B. Circulating DNA and myeloperoxidase indicate disease activity in
Med 17: 1381–1390, 2011.
patients with thrombotic microangiopathies. Blood 120: 1157–1164, 2012. 49. Pilsczek FH, Salina D, Poon KK, Fahey C, Yipp BG, Sibley CD,
27. Garcia-Romo GS, Caielli S, Vega B, Connolly J, Allantaz F, Xu Z, Robbins SM, Green FH, Surette MG, Sugai M, Bowden MG, Hussain
Punaro M, Baisch J, Guiducci C, Coffman RL, Barrat FJ, M, Zhang K, Kubes P. A novel mechanism of rapid nuclear neutrophil
Banchereau J, Pascual V. Netting neutrophils are major inducers of type extracellular trap formation in response to Staphylococcus aureus. J
I IFN production in pediatric systemic lupus erythematosus. Sci Transl Immunol 185: 7413–7425, 2010.
Med 3: 73ra20, 2011. 50. Reid VL, Gleeson M, Williams N, Clancy RL. Clinical investigation of
28. Gleeson M, Bishop NC, Stensel DJ, Lindley MR, Mastana SS, Nimmo athletes with persistent fatigue and/or recurrent infections. Br J Sports
MA. The anti-inflammatory effects of exercise: mechanisms and implica- Med 38: 42–45, 2004.
tions for the prevention and treatment of disease. Nat Rev Immunol 11: 51. Sangaletti S, Tripodo C, Chiodoni C, Guarnotta C, Cappetti B,
607–615, 2011. Casalini P, Piconese S, Parenza M, Guiducci C, Vitali C, Colombo
29. Guiducci C, Tripodo C, Gong M, Sangaletti S, Colombo MP, Coffman MP. Neutrophil extracellular traps mediate transfer of cytoplasmic neu-
RL, Barrat FJ. Autoimmune skin inflammation is dependent on plasma- trophil antigens to myeloid dendritic cells toward ANCA induction and
cytoid dendritic cell activation by nucleic acids via TLR7 and TLR9. J Exp associated autoimmunity. Blood 120: 3007–3018, 2012.
Med 207: 2931–2942, 2010. 52. Sengelov H, Kjeldsen L, Borregaard N. Control of exocytosis in early
30. Gunzer M. Traps and hyper inflammation—new ways that neutrophils neutrophil activation. J Immunol 150: 1535–1543, 1993.
promote or hinder survival. Br J Haematol 164: 189 –199, 2014. 53. Simon D, Simon HU, Yousefi S. Extracellular DNA traps in allergic,
31. Hakkim A, Furnrohr BG, Amann K, Laube B, Abed UA, Brinkmann infectious, and autoimmune diseases. Allergy 68: 409 –416, 2013.
V, Herrmann M, Voll RE, Zychlinsky A. Impairment of neutrophil 54. Swarup V, Rajeswari MR. Circulating (cell-free) nucleic acids—a prom-
extracellular trap degradation is associated with lupus nephritis. Proc Natl ising, non-invasive tool for early detection of several human diseases.
Acad Sci USA 107: 9813–9818, 2010. FEBS Lett 581: 795–799, 2007.
32. Holdenrieder S, Stieber P. Clinical use of circulating nucleosomes. Crit 55. Urban CF, Ermert D, Schmid M, bu-Abed U, Goosmann C, Nacken
Rev Clin Lab Sci 46: 1–24, 2009. W, Brinkmann V, Jungblut PR, Zychlinsky A. Neutrophil extracel-
33. Kahlenberg JM, Kaplan MJ. Mechanisms of premature atherosclerosis lular traps contain calprotectin, a cytosolic protein complex involved in
in rheumatoid arthritis and lupus. Annu Rev Med 64: 249 –263, 2013. host defense against Candida albicans. PLoS Pathog 5: e1000639,
34. Kessenbrock K, Krumbholz M, Schonermarck U, Back W, Gross WL, 2009.
Werb Z, Grone HJ, Brinkmann V, Jenne DE. Netting neutrophils in 56. Von Bruhl ML, Stark K, Steinhart A, Chandraratne S, Konrad I,
autoimmune small-vessel vasculitis. Nat Med 15: 623–625, 2009. Lorenz M, Khandoga A, Tirniceriu A, Coletti R, Kollnberger M,
35. Kraemer RR, Brown BS. Alterations in plasma-volume-corrected blood Byrne RA, Laitinen I, Walch A, Brill A, Pfeiler S, Manukyan D,
components of marathon runners and concomitant relationship to perfor- Braun S, Lange P, Riegger J, Ware J, Eckart A, Haidari S,
mance. Eur J Appl Physiol Occup Physiol 55: 579 –584, 1986. Rudelius M, Schulz C, Echtler K, Brinkmann V, Schwaiger M,
36. Lippi G, Maffulli N. Biological influence of physical exercise on hemo- Preissner KT, Wagner DD, Mackman N, Engelmann B, Massberg
stasis. Semin Thromb Hemost 35: 269 –276, 2009. S. Monocytes, neutrophils, and platelets cooperate to initiate and

J Appl Physiol • doi:10.1152/japplphysiol.00173.2014 • www.jappl.org


NETs in Exercise • Beiter T et al. 333
propagate venous thrombosis in mice in vivo. J Exp Med 209: 819 – CJ, Northoff H, Abbasi A, Simon P. Position statement. Part one:
835, 2012. immune function and exercise. Exerc Immunol Rev 17: 6 –63, 2011.
57. Walsh NP, Gleeson M, Pyne DB, Nieman DC, Dhabhar FS, Shephard 59. Yipp BG, Petri B, Salina D, Jenne CN, Scott BN, Zbytnuik LD,
RJ, Oliver SJ, Bermon S, Kajeniene A. Position statement. Part two: Pittman K, Asaduzzaman M, Wu K, Meijndert HC, Malawista SE, de
maintaining immune health. Exerc Immunol Rev 17: 64 –103, 2011. Boisfleury CA, Zhang K, Conly J, Kubes P. Infection-induced NETosis
58. Walsh NP, Gleeson M, Shephard RJ, Gleeson M, Woods JA, Bishop is a dynamic process involving neutrophil multitasking in vivo. Nat Med
NC, Fleshner M, Green C, Pedersen BK, Hoffman-Goetz L, Rogers 18: 1386 –1393, 2012.

Downloaded from on November 5, 2014

J Appl Physiol • doi:10.1152/japplphysiol.00173.2014 • www.jappl.org

You might also like