Embolismo de Critales de Colesterol

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Circulation Research

ORIGINAL RESEARCH

Crystal Clots as Therapeutic Target in


Cholesterol Crystal Embolism
Chongxu Shi, Tehyung Kim, Stefanie Steiger, Shrikant R. Mulay, Barbara M. Klinkhammer, Tobias Bäuerle,
Maria Elena Melica, Paola Romagnani, Diana Möckel, Maike Baues, Luying Yang, Sanne L.N. Brouns,
Johan W. M. Heemskerk, Attila Braun, Twan Lammers, Peter Boor,* Hans-Joachim Anders *

RATIONALE: Cholesterol crystal embolism can be a life-threatening complication of advanced atherosclerosis. Pathophysiology
and molecular targets for treatment are largely unknown.

OBJECTIVE: We aimed to develop a new animal model of cholesterol crystal embolism to dissect the molecular mechanisms of
cholesterol crystal (CC)–driven arterial occlusion, tissue infarction, and organ failure.

METHODS AND RESULTS: C57BL/6J mice were injected with CC into the left kidney artery. Primary end point was glomerular filtration
rate (GFR). CC caused crystal clots occluding intrarenal arteries and a dose-dependent drop in GFR, followed by GFR recovery
within 4 weeks, that is, acute kidney disease. In contrast, the extent of kidney infarction was more variable. Blocking necroptosis
using mixed lineage kinase domain–like deficient mice or necrostatin-1s treatment protected from kidney infarction but not from
GFR loss because arterial obstructions persisted, identifying crystal clots as a primary target to prevent organ failure. CC involved
platelets, neutrophils, fibrin, and extracellular DNA. Neutrophil depletion or inhibition of the release of neutrophil extracellular traps
had little effects, but platelet P2Y12 receptor antagonism with clopidogrel, fibrinolysis with urokinase, or DNA digestion with
recombinant DNase I all prevented arterial occlusions, GFR loss, and kidney infarction. The window-of-opportunity was <3 hours
after CC injection. However, combining Nec-1s (necrostatin-1s) prophylaxis given 1 hour before and DNase I 3 hours after CC
Downloaded from http://ahajournals.org by on May 8, 2023

injection completely prevented kidney failure and infarcts. In vitro, CC did not directly induce plasmatic coagulation but induced
neutrophil extracellular trap formation and DNA release mainly from kidney endothelial cells, neutrophils, and few from platelets.
CC induced ATP release from aggregating platelets, which increased fibrin formation in a DNase-dependent manner.

CONCLUSIONS: CC embolism causes arterial obstructions and organ failure via the formation of crystal clots with fibrin, platelets,
and extracellular DNA as critical components. Therefore, our model enables to unravel the pathogenesis of the CC embolism
syndrome as a basis for both prophylaxis and targeted therapy.

VISUAL OVERVIEW: An online visual overview is available for this article.

Key Words:  acute kidney injury ◼ endothelial cells ◼ extracellular traps ◼ fibrin ◼ necroptosis

In This Issue, see p 943 | Meet the First Author, see p 944

A
therosclerosis is a leading cause of global mor- Little is known about the precise cellular and molecular
bidity and mortality.1,2 In advanced atherosclerosis, mechanisms following CC embolism, in part, due to the
cholesterol crystal (CC) embolism is a potentially lack of animal models.11 We hypothesized that develop-
life-threatening complication with an average mortal- ing a reproducible mouse model of CC embolism would
ity of 62.8%.3–9 Autopsies or tissue biopsies reveal be instrumental to dissect the molecular mechanisms
CC inside the arterial lumen surrounded by an unde- of CC-driven arterial occlusion, tissue infarction, and
fined biological matrix obstructing the vessel lumen.7,10 organ failure.

Correspondence to: Hans-Joachim Anders, MD, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ziemssenstr. 1, 80336 München, Germany.
Email hjanders@med.uni-muenchen.de
*P.B. and H.-J.A. contributed equally to this article.
The Data Supplement is available with this article at https://www.ahajournals.org/doi/suppl/10.1161/CIRCRESAHA.119.315625.
For Sources of Funding and Disclosures, see page e50.
© 2020 American Heart Association, Inc.
Circulation Research is available at www.ahajournals.org/journal/res

Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625 April 10, 2020   e37


Shi et al Crystal Clots in Cholesterol Embolism

Novelty and Significance


Original Research

What Is Known? Cholesterol crystal embolism (CCE) is a potentially


• Cholesterol crystal embolism is a potentially life-threat- life-threatening complication of advanced atheroscle-
ening complication of advanced atherosclerosis. rosis, due to lack of animal models, we know very little
• Lack of reproducible animal model to investigate the about the precise cellular and molecular mechanisms
mechanism of cholesterol crystal embolism. following CCE. Our new animal model mimics the
• Platelet adhesion promotes thrombus growth. morphological and functional characteristics of CCE
• Extracellular DNA is an important component of vas- in human. We found that clots formed around the crys-
cular thrombosis. tals cause arterial obstruction and organ failure rather
than crystals themselves. Therefore, crystal clots are
What New Information Does This Article therapeutic and indeed targeting thrombosis and
Contribute? hemostasis, especially enhancing fibrinolysis or inhib-
• Established a new animal model to mimic the morpho- iting platelet purinergic signaling could reduce arterial
logical and functional characteristics of cholesterol occlusions and organ failure. Our results suggest that
crystal embolism in human. prophylactic necroptosis inhibition with a combination
• Not the crystal embolism but the clots forming around
of DNase I therapy could have a synergistic effect
the crystals cause arterial obstruction and organ failure.
on CC induced clot formation in mice and might be a
• Extracellular DNA is a critical element of crystal-induced
arterial occlusion and can be a target for therapy. feasible 2-step prophylactic/therapeutic approach in
• Combining preemptive necroptosis inhibition plus human with a risk for procedure-related CCE.
delayed DNase I treatment could be a feasible therapy
in patients at risk for cholesterol crystal embolism.

as assessed by post-interventional scoring (not shown),


as this way we avoided discomfort from skin ulcerations,
Nonstandard Abbreviations and Acronyms pancreatitis, peritonitis, or uremia. Injecting different
Downloaded from http://ahajournals.org by on May 8, 2023

amounts of CC resulted in a dose-dependent decline of


α-SMA α-smooth muscle actin glomerular filtration rate (GFR) at 24 hours, that is, acute
CC cholesterol crystal kidney injury (Figure 1A). Ex vivo magnetic resonance
CRP collagen-related peptide imaging displayed tissue defects and perilesional signal
ecDNA extracellular DNA enhancement inside kidneys (Figure 1B; Movie I in the
GFR glomerular filtration rate Data Supplement). The macroscopic analysis revealed
GPVI-ITAM glycoprotein VI-immunoreceptor kidney swelling and territorial kidney infarctions, with
tyrosine–based activation motif much higher dose-dependent infarct size variabilty
MLKL mixed lineage kinase domain–like compared with GFR (Figure 1C and 1D; Figure IC in
NLRP3 NOD-like receptor pyrin domain–con- the Data Supplement). Microscopic analysis revealed
taining protein 3 dose-dependent intravascular CC, wide areas of
PAR protease activated receptor TUNEL+ (TdT-mediated dUTP-biotin nick-end labeling)
corticomedullary necrosis, interstitial edema, neutrophil
infiltrates, and loss of CD31+ microvasculature (Fig-
METHODS ure 1E; Figure ID through IG in the Data Supplement).
The authors declare that all supporting data are available within Territorial infarctions were associated with complete or
the article (and in the Data Supplement). A detailed description partial occlusions of Aa. interlobares, Aa. Arcuatae, or
of the experimental procedures, data analysis, and statistical
Aa. interlobulares proximal to the infarct as assessed by
methods is provided in the Data Supplement. All methods have
corresponding literature references. α-SMA (α-smooth muscle actin)/fibrin staining to iden-
tify intrarenal arteries (Figure 1F through 1F′′; Figure
VIIIA in the Data Supplement). Three-dimensional (3D)
RESULTS reconstructions of arterial contrast-enhanced micro-
computed tomography angiography showed partial and
CC Injection Induces Arterial Occlusions, Tissue complete arterial occlusions with associated blood ves-
Infarction, and Organ Failure in C57BL/6J Mice sel rarefaction, blood vessel volume change, and vaso-
Injecting CC into the left kidney artery (Figure IA and IB constriction (Figure 1G through 1G′′; Movies II and III
in the Data Supplement) was reliable and well-tolerable in the Data Supplement). We selected the CC dose of

e38   April 10, 2020 Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625


Shi et al Crystal Clots in Cholesterol Embolism

Original Research
Downloaded from http://ahajournals.org by on May 8, 2023

Figure 1. A new model of cholesterol crystal (CC) embolism.


A, Injection of different doses of CC stock solution into the renal artery of C57BL/6J mice induced acute kidney injury as defined by a sudden
drop in glomerular filtration rate (GFR) 24 h post-injection (P=0.000000001 for Kruskal-Wallis test for whole group, followed by Dunn post-test).
B, Magnetic resonance imaging (MRI) indicates a large tissue defect, here in the renal medulla in the injected vs the noninjected contralateral
kidney (see also Movie I in the Data Supplement). C, Periodic acid-Schiff (PAS) stained section of control and CC embolism kidney. (Continued )

Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625 April 10, 2020   e39


Shi et al Crystal Clots in Cholesterol Embolism

10 mg/kg for further studies, which would allow the Neutrophils contribute to arterial thrombosis by releas-
detection of both disease improvement or aggravation. ing extracellular traps from neutrophils,18 a process
Original Research

Disease severity at 24 hours was identical in male and initiated by activated platelets.19,20 Surprisingly, crystal
female mice (Figure IH and I I in the Data Supplement). clots stained weakly positive for histological markers
Time-course analysis showed that after 14 days, GFR of neutrophils including extracellular DNA (ecDNA),
recovered back to baseline and infarct size declined citrullinated histone H3, and cytoplasmic proteins such
(Figure IJ and IK in the Data Supplement). Thus, intra- as elastase or granular proteins such as myeloper-
arterial CC injection induces arterial occlusions causing oxidase21,22 (Figure 3E). Eosinophils were not found
acute territorial infarctions, perilesional inflammation, (not shown). To further test the potential contribution
and organ failure. of neutrophils to crystal clot formation, we depleted
neutrophils with anti-Ly6G IgG (Figure IIIA in the
Data Supplement) as confirmed by blood flow cytom-
Blocking Necroinflammation Prevents Kidney etry done before CC injection (Figure IIIB in the Data
Infarction but not Kidney Failure Supplement). At 24 hour after CC injection, neutrophil
MLKL (mixed lineage kinase domain–like)-dependent depletion had completely extinguished the periinfarct
necroptosis and NLRP3 (NOD-like receptor pyrin neutrophil infiltrates and significantly decreased kid-
domain–containing protein 3) inflammasome-depen- ney infarct size compared with IgG control group, but
dent sterile inflammation contribute to tubular necro- this had no significant effect on arterial obstructions
sis and loss of kidney function upon transient artery or GFR loss, maybe because mononuclear cells had
clamping.13–17 Indeed, injecting 10 mg/kg CC into the partially replaced neutrophils in the crystal clots, and
renal arteries of Mlkl-deficient mice, mice pretreated ecDNA was still present in crystal clots of the neutro-
with the necroptosis inhibitor Nec-1s (necrostatin-1s) phil depletion group (Figure 3F through 3I′; Figures IIIF
or the NLRP3 inhibitor MCC950 significantly reduced and IIIF′ and IXA in the Data Supplement). As another
infarct size, kidney injury, and neutrophil infiltration at approach, Cl-amidine was used to block PAD4 (pepti-
24 hours compared with wild-type controls (Figure 2A dylarginine deiminase)-dependent neutrophil formation
through 2C). However, none of these interventions had with similar results (Figure IIIG through IIIJ′ and Figure
an effect on CC embolism–related loss of GFR (Fig- IXB in the Data Supplement). In contrast, the platelet
ure 2E). Perfusion of the viable nephrons is a central P2Y12 receptor antagonist clopidogrel completely pro-
Downloaded from http://ahajournals.org by on May 8, 2023

determinant of kidney excretory function, and consis- tected mice from intravascular obstructions, GFR loss,
tently with GFR, interfering with CC embolism–induced kidney infarction, and perilesional neutrophil infiltrates
necroinflammation had no impact on arterial occlusions (Figure IIIC and Figure 3F through 3I′). Together, CC
at 24 hours (Figure 2D and 2D′ and Figure VIIIB in the occlude arteries by forming crystal clots consisting of
Data Supplement). Thus, as nephron perfusion is ulti- fibrin, platelets, and neutrophils, of which platelets but
mately required for kidney function, inhibiting necrosis not neutrophils are central for CC embolism–related
without targeting arterial occlusions does not prevent arterial occlusion, organ failure, and tissue infarction.
kidney failure.
CC Occlude Arteries by Forming Crystal Clots—
CC Occlude Arteries by Forming Crystal Clots— Role of Anticoagulants
Role of Neutrophils and Platelets The fibrin mesh is a validated target for arterial and
Interestingly, CC were a minor component of vascu- venous thrombosis, and fibrin was also present in crys-
lar occlusions, while vascular obstruction was rather tal clots. Hence, we tested the effects of the antico-
related to the surrounded material staining positive for agulant heparin and the fibrinolytic agent urokinase
fibrin, CD61+ platelets, and Ly6B2+ neutrophils (Fig- (Figure IVA in the Data Supplement). At 24 hours, both
ure 3A through 3D), the typical components of arte- heparin and urokinase significantly reduced the num-
rial thrombi.3 Therefore, we named them crystal clots. bers of arterial occlusions, albeit the crystal component

Figure 1 Continued. C′, Infarct (*) demarcation (tubular damage, bleeding, neutrophils). C′′, Diffuse (tubular) ischemic damage and arterial
crystal (*). D, Triphenyl tetrazolium chloride (TTC)-stained kidney slices allowed to distinguish viable from infarcted tissue and calculation of
infarct area (P=0.000000001 for Kruskal-Wallis test for whole group, followed by Dunn post-test). E, CC-injected kidney showing crystal
inside artery. F, Schematic depiction of kidney vasculature. F′, Immunostaining for α-SMA (α-smooth muscle actin) and fibrin displays partial
or complete arterial occlusions. Arrows pointing to CC. F′′, Quantitative results for obstructed intrarenal arteries of different sizes. We set 6
different parameters to analyze the ratio of obstructed arteries (Details see Figure VIIIA in the Data Supplement): partially and completely
interlobar, partially and completely arcuate, partially and completely interlobular. F′′ only shown the mean value of each parameters in each
group. G, Three-dimensional reconstructions of arterial contrast micro computed tomography displays peripheral vascular rarefication (Movies
II and III in the Data Supplement), G′, Blood vessel volume change in CCE group. G′′, CCE induced vasoconstriction. All quantitative data are
means±SD. G′ and G′′ used t test.

e40   April 10, 2020 Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625


Shi et al Crystal Clots in Cholesterol Embolism

Original Research
Downloaded from http://ahajournals.org by on May 8, 2023

Figure 2. Role of necroinflammasome in cholesterol crystal (CC) embolism.


We injected 10 mg/kg CC stock solution into the renal arteries of wild type (WT), Mlkl−/− mice on C57BL/6J background. A, Infarct size was
determined from 2,3,5-triphenyltetrazolium chloride (TTC)-stained kidney slices and shown as percentage of kidney mass in WT, Mlkl−/−, Nec-1s
(necrostatin-1s), and MCC950 treatment groups. All those treatments significantly reduced kidney infarct size (P=0.000000055 for Kruskal-Wallis
test for whole group, followed by Dunn post-test). B, Representative periodic acid-Schiff (PAS) stained sections of CC-injected kidneys in WT, Mlkl−/−
mice, Nec-1s or MCC950 treated groups, arrows show tubular damage. Mlkl−/− mice, Nec-1s or MCC950 treated groups show significantly less injury
(P=0.00000008 for Kruskal-Wallis test for whole group, followed by Dunn post-test). C, Perilesional neutrophil infiltrates; * indicates crystals. Mlkl−/−
mice and treated groups had less neutrophils (P=0.0003 for Kruskal-Wallis test for whole group, followed by Dunn post-test). D, Representative α-
SMA (α-smooth muscle actin)/Fibrin stained section. D′, Occlusions of renal arteries of various sizes, same analysis as Figure 1F′′ (details see Figure
VIIIB in the Data Supplement). E, Glomerular filtration rate (GFR) was measured at 24 hour in WT, Mlkl−/− mice, Nec-1s, or MCC950 treated groups
to assess any drop compared to baseline GFR, an indicator of kidney failure. Mlkl−/− mice and treated groups had no effects on kidney function
(P=0.00000004 for Kruskal-Wallis test for whole group, followed by Dunn post-test). All quantitative data are means±SD.

Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625 April 10, 2020   e41


Shi et al Crystal Clots in Cholesterol Embolism
Original Research
Downloaded from http://ahajournals.org by on May 8, 2023

Figure 3. Components of intraarterial crystal clots (CC)—role of neutrophils and platelets.


A, Periodic acid-Schiff (PAS) staining shows that intravascular crystals are surrounded by a biological mass. Immunostaining of crystal clots
with prominent positivity CC for fibrinogen (B), platelets (C), and neutrophils (D). E, neutrophil extracellular trap (NET) formation illustrated
by extracellular positivity of citrullinated histone H3 and MPO (myeloperoxidase). F, Representative image of Ly6B2+ stained kidney section
in control, anti-Ly6G and clopidogrel groups, neutrophil depletion, and platelet antagonist significantly reduced neutrophil infiltration into the
kidney (P=0.0000043 for Kruskal-Wallis test for whole group, followed by Dunn post-test). G, Neutrophil depletion had no significant effect on
CC injection-induced loss of glomerular filtration rate (GFR; P=0.00000065 across groups, 1-way ANOVA, followed by Dunnett post-test). H,
Neutrophil depletion significantly reduced infarct size (P=0.000000009 for Kruskal-Wallis test for whole group, followed by Dunn post-test),
and platelet antagonist had a complete protection on GFR loss (G) and infarct size (H). The latter indicated by the percentage of kidney mass
as compared with mice injected with control IgG. I, Representative αSMA (α-smooth muscle actin)/fibrin stained section in neutrophil depletion
and platelet inhibition group. I′, Occlusions of renal arteries of various sizes in neutrophil depletion and platelet inhibition group. Same analysis as
Figure 1F′′ (details see Figure IXA in the Data Supplement). All quantitative data are means±SD.

e42   April 10, 2020 Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625


Shi et al Crystal Clots in Cholesterol Embolism

Original Research
Downloaded from http://ahajournals.org by on May 8, 2023

Figure 4. Anticoagulants protect from tissue infarction and organ failure.


A: Representative α-SMA (α-smooth muscle actin)/fibrin stained section in heparin and urokinase treatment groups, A′: occlusion of renal
arteries of varies sizes were identified and quantified. Same analysis as Figure 1F′′ (details see Figure XA in the Data Supplement). Heparin and
urokinase had both profound protective effects on cholesterol crystal (CC) injection-induced infarct size (B; P=0.00000018 for Kruskal-Wallis
test for whole group, followed by Dunn post-test), loss of glomerular filtration rate (GFR; C, P=0.00000207 for Kruskal-Wallis test for whole
group, followed by Dunn post-test), and the number of TUNEL+ (TdT-mediated dUTP-biotin nick-end labeling) dying cells (D, P=0.0013 for
Kruskal-Wallis test for whole group, followed by Dunn post-test). E, Representative Periodic acid-Schiff (PAS) stained sections in heparin and
urokinase treatment groups and kidney injury score, arrows show tubular damage; * indicates crystals. All quantitative data are means±SD. DAPI
indicates 4′,6-diamidino-2-phenylindole; and TCC, 2,3,5-triphenyltetrazolium chloride.

persisted (Figure 4A and 4A′; Figure XA in the Data infarct size, kidney injury, neutrophils infiltration, vascu-
Supplement). This was associated with complete pro- lar injury as well as kidney cell death as indicated by
tection from GFR loss, a significant reduction in kidney TUNEL positivity compared with vehicle-treated mice

Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625 April 10, 2020   e43


Shi et al Crystal Clots in Cholesterol Embolism
Original Research
Downloaded from http://ahajournals.org by on May 8, 2023

Figure 5. Neutralization of extracellular DNA protects mice from tissue infarction and organ failure.
A, Representative DAPI (4′,6-diamidino-2-phenylindole)-stained sections in cholesterol crystal embolism (CCE) kidneys showing crystals clots
without and with extracellular DNA (ecDNA; arrows). B, Quantification of arteries containing crystal clots with ecDNA in healthy, recombinant
DNase I or NaCl treated mice kidney after CC injection. DNase I treatment significantly decreased the percentage of arteries with ecDNA
positive clots (P=0.0002 for Kruskal-Wallis test for whole group, followed by Dunn post-test). C, Occlusions of renal arteries of varying sizes were
identified and quantified. Recombinant DNase I significantly reduced the number of completely or partially obstructed interlobar, arcuate, and
interlobular arteries. We set 6 different parameters to analyze the ratio of obstructed artery (Figure XB in the Data Supplement): (Continued )

e44   April 10, 2020 Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625


Shi et al Crystal Clots in Cholesterol Embolism

(Figure 4B through 4E; Figure IVB and IVC in the Data MCC950, or Cl-amidine had no effect on DNA release
Supplement). Conclusively, not the crystals per se but (Figure VIC and VID in the Data Supplement). Exposure

Original Research
rather crystal clots cause arterial obstruction, tissue of human platelets to CC also induced the release of
infarction, and organ failure. minor amounts of ecDNA from their mitochondria (Fig-
ure VIE in the Data Supplement). Thus, in vitro studies
support that CC and platelet-dependent ecDNA release
CC Occlude Arteries by Forming Crystal Clots— from neutrophils and endothelial cells could be a contrib-
Role of Extracellular DNA uting factor in CC-induced arterial obstruction.
Accumulation of ecDNA in the fibrin clot can be an impor-
tant component of vascular thrombosis.21,23 Using DAPI
(4′,6-diamidino-2-phenylindole) staining, we noticed
DNase I Abrogates CC-Induced Platelet
positivity for ecDNA in crystal clots (Figure 5A) and Activation and Fibrin Formation
recombinant DNase I treatment significantly decreased Given the efficacy of clopidogrel in inhibiting crystal clot
the percentage of CC clots (Figure 5B). To test its func- formation, we considered that ATP release and puriner-
tional contribution for arterial occlusions, we treated gic receptor P2Y12 signaling are directly involved in
C57BL/6J mice with recombinant DNase I (Figure VA in this process.24 We exposed washed mouse platelets to
the Data Supplement). After 24 hours of DNase I treat- thrombin in the presence or absence of CC and quanti-
ment, intraarterial ecDNA had disappeared together with fied fibrin formation with a turbidity assay at 405 nm. CC
a reduction in the number of arterial occlusions, and the did not influence turbidity in resting platelets, but upon
fibrin and cellular components of crystal clots, while the thrombin activation, turbidity decreased in CC-treated
crystal component, persisted (Figure 5C and 5C′; Figure platelets (Figure 6J). Thus, CC enhances fibrinogen
XB in the Data Supplement). This effect was confirmed release from platelet alpha (α)-granules, which further
by 3-dimensional micro-computed tomography imaging promotes fibrin clot formation. CC exposure also induced
(Figure 5D; Movies IV and V in the Data Supplement). ATP secretion from dense (δ)-granules, but co-incuba-
Preventing arterial occlusions was associated with a tion with DNase I strongly reduced these extracellular
complete protection from GFR loss, a significant reduc- ATP levels to the levels of untreated platelets (Fig-
tion in kidney infarct size as well as kidney cell death as ure 6K). Next, we stimulated platelets with thrombin and
indicated by less TUNEL positivity, neutrophil infiltrates, CRP (collagen-related peptide) to activate PAR-Gq (pro-
and vascular rarefaction (Figure 5E through 5G; Figure
Downloaded from http://ahajournals.org by on May 8, 2023

tease activated receptor) and GPVI-ITAM (glycoprotein


VB through VD in the Data Supplement). Thus, ecDNA VI-immunoreceptor tyrosine-based activation motif) sig-
is another nonredundant component of CC embolism– naling, respectively, and degranulation was monitored by
related arterial obstruction, tissue infarction, and organ an α-granule marker P-selectin. In the presence of CC, a
failure. significant increase of P-selectin exposure was detected,
while DNase I treatment could inhibit this process in the
presence or absence of CC (Figure 6L and 6M). This
CC Directly and Indirectly Induce DNA Release indicated a crucial role of ecDNA in platelet degranula-
From Several Cellular Sources tion, thereby inhibiting fibrinogen and ATP secretion and
Because DNase I treatment was superior to neutrophil subsequent fibrin formation and P2Y12 receptor signal-
depletion in protecting the kidney, ecDNA should derive ing, respectively.
from multiple sources. We addressed this by a series Finally, a known element in local platelet adhesion
of in vitro studies. Exposing CC or supernatant of CC- and thrombus growth in CC embolism is vascular col-
activated platelets to human neutrophils induced neu- lagen matrix.25 To model this process in vitro, we tested
trophil necrosis and neutrophil-like chromatin release collagen-driven platelet aggregation in the presence
and free DNA in the cell-culture supernatant (Figure 6A or absence of CC. Results showed that collagen I trig-
through 6C). Exposure to increasing doses of CC also gered massive platelet aggregation in 5 minutes in the
induced renal endothelial cells to undergo necrosis and presence of CC, and DNase I treatment normalized this
to release DNA in 2D as well as 3D culture (Figure 6D accelerated aggregation response (Figure 6N and 6O).
through 6I, Figure VIA and VIB and Movies VI and VII Thus, DNase I can attenuate CC-induced platelet activa-
in the Data Supplement). Pretreated GEnC with Nec-1s, tion, aggregation, and fibrin clot formation.

Figure 5 Continued. partially and completely interlobar, partially and completely arcuate, partially and completely interlobular. C′ only shown
the mean value of each parameters in each group. D, Three-dimensional micro-computed tomography of CC-injected kidneys confirmed the
attenuation of vascular occlusions by a higher density of microvascular perfusion. Right, The foremost artery branch outlined by the dashed
rectangle in the Left panel (see also Movies IV and V in the Data Supplement). E/F, Recombinant DNase I had profound protective effects on CC
injection-induced loss of glomerular filtration rate (GFR; E, P=0.000045 across groups, 1-way ANOVA, followed by Dunnett post-test), infarct size
(F, P=0.0000054 for Kruskal-Wallis test for whole group, followed by Dunn post-test), and the number of TUNEL+ (TdT-mediated dUTP-biotin
nick-end labeling) dying cells (G, P=0.0003 for Kruskal-Wallis test for whole group). All quantitative data are means±SD.

Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625 April 10, 2020   e45


Shi et al Crystal Clots in Cholesterol Embolism
Original Research
Downloaded from http://ahajournals.org by on May 8, 2023

Figure 6. In vitro studies with endothelial cells, platelets, and neutrophils.


A–C, Supernatants of platelets exposed to cholesterol crystal–induced neutrophil extracellular trap (NET) formation in neutrophils (A, confocal
microscopy with Sytox Green ecDNA staining), cell death (B, P=0.000000001 across groups, 1-way ANOVA, followed by Dunnett post-test) and
DNA release (C, P=0.000000001 across groups, 1-way ANOVA, followed by Dunnett post-test) of human neutrophil. D–F, Cholesterol crystal
(CC) induced mouse glomerular endothelial cells to release DNA (D, confocal microscopy with Sytox Green, P=0.000000001 across groups,
1-way ANOVA, followed by Dunnett post-test), to undergo cell death (E, P=0.000000001 across groups, 1-way ANOVA, followed by Dunnett
post-test), and to release DNA into the cell culture supernatant, where it was measured by Pico green Kit (Continued )

e46   April 10, 2020 Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625


Shi et al Crystal Clots in Cholesterol Embolism

A 2-Step Strategy to Improve Outcomes of CC but our data also provide a new pathophysiological
Embolism concept for CC embolism (Figure 8) and identify novel

Original Research
molecular targets for prophylaxis and therapy.
As cardiac or aorta surgeries preclude the use of anti- This inducible model of dose-dependent CC embo-
coagulants or fibrinolytic agents, we considered recom- lism in male and female mice mimics the morpho-
binant DNase I as a possible alternative to attenuate logical and functional characteristics of CC embolism
CC clot formation by inhibiting fibrin formation, ecDNA syndrome in humans,7,10 rabbits, and rats.27–30 Different
accumulation, and ATP signaling. First, we tested the ways of CC preparation may result in CC of different
therapeutic window-of-opportunity and administered shapes, that is, plate or needle; however, morphologi-
recombinant DNase I 3, 6, and 12 hours after CC injec- cally the shape of crystals found in our model were
tion (Figure VIIA in the Data Supplement). While DNase identical to those found in patients. Still, the ex vivo CC
I given at 6 and 12 hours lacked effects on any of the preparation may differ from the lipid material dislocat-
end points, DNase I treatment given 3 hours after CC ing in human diseases. Organ-specific effects have to
embolism showed trends toward improved outcomes be kept in mind as rapid spontaneous revasculariza-
albeit not in a consistent manner (Figure 7A through tion was reported selectively for pulmonary artery CC
7D; Figure VIIB through VIID in the Data Supplement). embolism and rabbit muscle arteries can lack crystal
To further optimize outcomes in the setting of a car- clot formation.27,31 Functionally, in our model, the GFR
diovascular procedure-related CC embolism, we tested normalized within 14 days, most likely due to compen-
a regimen combining a preemptive single dose of the satory hypertrophy of the contralateral kidney and the
necroptosis inhibitor Nec-1s with therapeutic recombi- nonaffected parts of the cholesterol crystal embolism
nant DNase I given 3 hours after intraarterial CC injec- kidney. In contrast to previous attempts,27–31 our model
tion (Figure VIIE in the Data Supplement). This approach allows functional studies in genetically modified mice.
could be feasible as a prophylaxis given to all patients Specifically abrogating necroptosis, a form of regulated
at risk, while DNase I would be only given to those with necrosis well-known to mediate post-ischemic kidney
signs of CC embolism into the kidney, for example, necrosis,13–16,32,33 as well as NLRP3-dependent sterile
an early decline of urinary output. This dual strategy inflammation, that is, necroinflammation, prevented CC
resulted in a significant protection from GFR loss and embolism-induced kidney infarction, but not kidney fail-
kidney infarction in almost all animals together with a ure. This dissociation between tissue viability and func-
significant reduction in vascular occlusions by crystal
Downloaded from http://ahajournals.org by on May 8, 2023

tion was already obvious from the different variabilities


clots (Figure 7E through 7G′; Figure VIIF through VIIH of CC embolism-induced GFR loss versus infarct size.
in the Data Supplement). This is because, in contrast to other organs, kidney
function directly depends on the perfusion of arteries
afferent to the glomerular filters, while kidney infarction
DISCUSSION depends on numerous other factors such as collateral
Crystal induced diseases involve shared and unique perfusions of tubules, inflammation, and complex stress
pathomechanisms.26 We had hypothesized that develop- responses contributing to kidney cell death. Indeed, this
ing a reproducible model of CC embolism would help to finding confirms our choice for GFR as primary end
dissect the pathophysiology underlying CC embolism- point and infarct size as only a secondary end point in
driven arterial occlusion, tissue infarction, and organ fail- the clinically relevant acute kidney injury context.
ure. Indeed, our novel mouse model not only mimics all In 1973, Warren and Vales28 reported human athero-
local aspects of human atheroembolic kidney disease, matous plaque material injections into rabbit kidneys

Figure 6 Continued. (F, P=0.000000001 across groups, 1-way ANOVA, followed by Dunnett post-test). G–I, Three-dimensional cultured
of human endothelial cells in a Mimetas kidney-on-the-chip system under low-flow conditions were not exposed (G) or exposed (H) to CC
(see also Movies VI and VII in the Data Supplement). CC induced propidium iodide (PI) positivity as a marker of cell death, while intact living
cells were stained with calcein (green). I, DNA release from human endothelial cells into the luminal from 12 independent experiments was
measured by Pico green Kit (P=0.0001, followed by Dunnett post-test). J, Turbidity assay on resting and thrombin-activated platelets. C57BL/6J
wild-type mouse platelets were activated with 0.1 U/mL thrombin in the presence or absence of 2.5 mg/mL CC. Representative scanning
electron microscopy images of fibrin clots in the presence and absence of CC, and turbidity was measured at 405 nm in an ELISA reader
(P=0.000000002 across groups, 1-way ANOVA, followed by Dunnett post-test). K, Relative levels of ATP released from mouse platelets
incubated in the presence or absence of 2.5 mg/mL CC and 10 U/mL DNase I (P=0.0000035 across groups, 1-way ANOVA, followed by
Dunnett post-test). L–M, Flow cytometric analysis of P-selectin exposure in mouse platelets upon stimulation with the indicated agonists in
the presence or absence of CC and DNase I (L: P=0.000036, M: P=0.0000001 across groups, 2-way ANOVA, followed by Bonferroni post-
test). Washed mouse platelets were stimulated with collagen I (5 µg/mL), and platelet aggregation was measured in the presence or absence
of cholesterol and DNase I with light transmission aggregometry and recorded for 5 min. N, Representative aggregation curves, O: Bar graphs
showing relative maximal aggregation of platelets (%) in response to collagen I (P=0.0000057 across groups, 1-way ANOVA, followed by
Dunnett post-test). CRP indicates collagen related peptide; HMVEC, human microvascular endothelial cell; LDH, lactate dehydrogenase; MFI,
mean fluorescence intensity; and SEM, scanning electron microscopy; and Thr, thrombin.

Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625 April 10, 2020   e47


Shi et al Crystal Clots in Cholesterol Embolism
Original Research
Downloaded from http://ahajournals.org by on May 8, 2023

Figure 7. A 2-step strategy prevents cholesterol crystal embolism (CCE)-induced tissue infarction and organ failure.
A–D, Recombinant DNase I treated on mice after CC injection different time points effect on glomerular filtration rate (GFR) loss (A,
P=0.000084 for Kruskal-Wallis test for whole group, followed by Dunn post-test), infarct size (B, P=0.0000081 for Kruskal-Wallis test
for whole group, followed by Dunn post-test), vascular occlusions (C, details see Figure XIA in the Data Supplement), and kidney injury (D,
P=0.00000045 for Kruskal-Wallis test for whole group, followed by Dunn post-test). E–G, Dual therapy involving preemptive treatment
with Nec-1s+recombinant DNase I given 3 h after CC injection significantly protected from GFR loss (E, P=0.00000076 for Kruskal-Wallis
test for whole group, followed by Dunn post-test), infarct size (F, P=0.000017 for Kruskal-Wallis test for whole group, followed by Dunn
post-test), and vascular occlusions (G). G′, Ratio of obstructed artery (Details see Figure XIB in the Data Supplement). All quantitative data
are means±SD.

e48   April 10, 2020 Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625


Shi et al Crystal Clots in Cholesterol Embolism

Original Research
Downloaded from http://ahajournals.org by on May 8, 2023

Figure 8. Human tissue and schematic illustration of cholesterol crystal (CC) embolism and its therapeutic targets.
A, Periodic acid-Schiff (PAS) staining of human kidney and spleen obtained from autopsy tissue of a patient that died shortly after cholesterol
crystal embolism (CCE). Crystal clefts (shown by arrows) indicate the presence of crystals in a thrombotic mass. B, Feulgen stained human kidney
and spleen. Extracellular DNA (ecDNA) is indicated by arrows. C, Inside arterial vessel. D, CC triggers the formation of crystal clots composed of
activated platelets, neutrophils, fibrin, and extracellular DNA. E, Targeting this crystal clot with either clopidogrel, heparin, uPA inhibitor (urokinase),
or recombinant DNase I can resolve the clot component. As the remaining crystal component alone does not account for a relevant vascular
obstruction, such interventions can prevent tissue infarction and organ failure.

Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625 April 10, 2020   e49


Shi et al Crystal Clots in Cholesterol Embolism

and brains and found thrombotic material around the amounts of CC cannot damage the endothelial cell
embolus.28–30 Our studies extend these observations by layer, and therefore, CC not always induces fibrin clot
Original Research

showing that CC not only triggers diffuse clot formation formation and endovascular obstruction.31 However, it
but also the crystal clot and not the CC emboli alone has been shown that CC can attack endothelial cells in
account for vascular occlusions, infarct, and kidney fail- various ways including complement activation, plasma
ure. This finding is important as it renders the forming membrane destabilization, and Src homology region 2
clot as putative target for therapeutic intervention to domain-containing phosphatase-2 signaling45–49 but
improve outcomes. our data suggest that necroptosis, inflammasome or
Current recommendations for the management PAD4-dependent pathways are not involved.
of patients with CC embolism rely on retrospective In summary, not CC by itself but the fibrin clots form-
reports of single case or small single-center patient ing around CC obstruct peripheral arteries causing tis-
series; no randomized controlled interventional trials sue infarction and organ failure. Hence, crystal clots
have thus far performed.5,7,34 Partially contradictory represent the primary target for therapeutic interven-
outcomes have been reported for the use of ste- tions. Among the possible molecular targets in thrombo-
roids or anticoagulants, probably because beyond the sis and hemostasis, especially enhancing fibrinolysis or
peripheral lesions, outcomes also depend on the sta- inhibiting platelet purinergic signaling could reduce arte-
bility of aortic plaques, intraplaque hemorrhages, and rial occlusions, infarction, and organ failure albeit with
the risk for repetitive episodes.8,34–39 Our model allows a relatively short window-of-opportunity up to 3 hours.
dissecting these 2 different aspects of pathophysiol- Our results suggest that prophylactic necroptosis inhibi-
ogy, and it clearly demonstrates that using a platelet tion with a combination of DNase I therapy could have a
antagonist or anticoagulants, by interfering with crys- synergistic effect on CC induced clot formation in mice
tal clot formation, can prevent peripheral tissue necro- and might be a feasible 2-step prophylactic/therapeutic
sis and organ failure. This, however, does not exclude approach in human patients with a risk for procedure-
effects on plaques to potentially impact on outcomes related CC embolism.
the opposite way.
In this context, our data on the role of ecDNA in
crystal clot-related arterial obstructions are of potential ARTICLE INFORMATION
interest. ecDNA has recently been identified as a criti- Received June 28, 2019; revision received February 5, 2020; accepted February
20, 2020.
cal component of thrombosis, either by employing mice
Downloaded from http://ahajournals.org by on May 8, 2023

mutant for DNases or by using recombinant DNase Affiliations


I.21,23,40,41 These studies documented neutrophils as From the Medizinische Klinik und Poliklinik IV, Klinikum der Universität, LMU
München, Germany (C.S., T.K., S.S., S.R.M., L.Y., H.-J.A.); Department of Nephrol-
the source of ecDNA. We considered the same source
ogy, Institute of Pathology (B.M.K, P.B.) and Institute for Experimental Molecular
also for crystal clots, but neutrophil depletion or PAD4 Imaging (D.M., M.B., T.L.), RWTH Aachen University Hospital, Germany; Preclinical
inhibition had little effects on vascular obstructions Imaging Platform Erlangen, Institute of Radiology, Friedrich-Alexander-Universität
and acute kidney injury compared with DNase I injec- Erlangen-Nürnberg, Germany (T.B.); Excellence Centre for Research, Transfer
and High Education for the development of DE NOVO Therapies (M.E.M., P.R.)
tion, probably as due to the rather small contribution of and Department of Experimental and Clinical Biomedical Sciences “Mario Serio”
neutrophils to intravascular DNA release in our model. (M.E.M., P.R.), University of Florence, Italy; Nephrology and Dialysis Unit, Meyer
Although also platelets could be a source of ecDNA Children’s University Hospital, Florence, Italy (P.R.); Department of Biochemistry,
CARIM, Maastricht University, The Netherlands (S.L.N.B., J.W.M.H.); and Walther-
release from their mitochondria,42,43 this process may Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians University
have a minor role in CC-induced fibrin clot formation Munich, German Center for Lung Research, Germany (A.B.).
in our in vivo mouse model. Surprisingly, DNase I can
Acknowledgments
inhibit platelet degranulation and ATP release, and James. M. Murphy and Warren Alexander, Cell Signalling and Cell Death Division,
strongly inhibits CC- and collagen-induced aggrega- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia supplied
tion responses, which may be independent from its the Mlkl−/− mice. We thank Dr Elmina Mammadova-Bach, Institute of Experimental
Biomedicine, University Hospital and Rudolf Virchow Center, University of Würz-
effects on ecDNA. Whether DNase I directly hydro- burg, Germany, for her support with platelet experiments.
lyzes ATP and thereby inhibits purinergic signaling in
platelets and other cell types remains to be studied. Sources of Funding
This work was funded by the Deutsche Forschungsgemeinschaft (AN372/16-
As another mechanism, CC directly damage the sur- 2, 20-2, 24-1) to H.-J. Anders, SFB/TRR57 to T. Lammers, and SFB/TRR57,
face of endothelial cells in vitro and ex vivo.44 Indeed, SFB/TRR219, BO3755/3-1, and BO3755/6-1 to P. Boor, and 374031971-TRR
our data suggest that endothelial cell injury could be 240/A09 to A. Braun. The German Ministry of Education and Research (BMBF:
STOP-FSGS-01GM1901A) supported P. Boor. The European Research Coun-
the major source of locally accumulated ecDNA within
cil (ERC) under the European Union’s Horizon 2020 research and innovation
the CC fibrin clot, rather than DNA released by neu- programme supported P. Romagnani (grant agreement number 648274) and
trophils or platelets. Although we could detect CC and T. Lammers (grant agreement number 309495). The Ministero dell’Istruzione,
dell’Università e della ricerca, PRIN 2017 supported P. Romagnani.
platelet-dependent neutrophil formation in vitro, the
origin of DNA was difficult to ultimately prove in our Disclosures
in vivo mouse model. It is important to note that small None.

e50   April 10, 2020 Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625


Shi et al Crystal Clots in Cholesterol Embolism

Supplemental Materials 20. Nakazawa D, Marschner JA, Platen L, Anders HJ. Extracellular traps
in kidney disease. Kidney Int. 2018;94:1087–1098. doi: 10.1016/j.
Major Resources Tables

Original Research
Expanded Materials and Methods kint.2018.08.035
Online Figures I–XI 21. Laridan E, Denorme F, Desender L, François O, Andersson T, Deckmyn H,
Online Videos I–VII Vanhoorelbeke K, De Meyer SF. Neutrophil extracellular traps in ischemic
Supplemental References50–59 stroke thrombi. Ann Neurol. 2017;82:223–232. doi: 10.1002/ana.24993
22. Nakazawa D, Kumar S, Desai J, Anders HJ. Neutrophil extracellular traps
in tissue pathology. Histol Histopathol. 2017;32:203–213. doi: 10.14670/
HH-11-816
REFERENCES 23. Jiménez-Alcázar M, Rangaswamy C, Panda R, Bitterling J, Simsek YJ, Long
1. GBD 2017 Causes of Death Collaborators.Global, regional, and national AT, Bilyy R, Krenn V, Renné C, Renné T, et al. Host DNases prevent vascular
age-sex-specific mortality for 282 causes of death in 195 countries and occlusion by neutrophil extracellular traps. Science. 2017;358:1202–1206.
territories, 1980-2017: a systematic analysis for the global burden of dis- doi: 10.1126/science.aam8897
ease study 2017. Lancet. 2018;392:1736–1788. doi: 10.1016/S0140- 24. Golebiewska EM, Poole AW. Platelet secretion: from haemostasis to
6736(18)32203-7 wound healing and beyond. Blood Rev. 2015;29:153–162. doi: 10.1016/j.
2. Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic blre.2014.10.003
options. Nat Med. 2011;17:1410–1422. doi: 10.1038/nm.2538 25. Wang N, Tall AR. Cholesterol in platelet biogenesis and activation. Blood.
3. Jackson SP. Arterial thrombosis–insidious, unpredictable and deadly. Nat 2016;127:1949–1953. doi: 10.1182/blood-2016-01-631259
Med. 2011;17:1423–1436. doi: 10.1038/nm.2515 26. Mulay SR, Anders HJ. Crystallopathies. N Engl J Med. 2016;374:2465–
4. Moolenaar W, Lamers CB. Cholesterol crystal embolization in the Nether- 2476. doi: 10.1056/NEJMra1601611
lands. Arch Intern Med. 1996;156:653–657. 27. Gore I, Mccombs HL, Lindquist RL. Observations on the fate of cho-
5. Agrawal A, Ziccardi MR, Witzke C, Palacios I, Rangaswami J. Cholesterol lesterol emboli. J Atheroscler Res. 1964;4:527–535. doi: 10.1016/
embolization syndrome: an under-recognized entity in cardiovascular inter- s0368-1319(64)80055-7
ventions. J Interv Cardiol. 2018;31:407–415. doi: 10.1111/joic.12483 28. Warren BA, Vales O. The ultrastructure of the stages of atheroembolic
6. Thadhani RI, Camargo CA Jr, Xavier RJ, Fang LS, Bazari H. Atheroem- occlusion of renal arteries. Br J Exp Pathol. 1973;54:469–478.
bolic renal failure after invasive procedures. Natural history based on 52 29. Warren BA, Vales O. The ultrastructure of the reaction of arterial walls to
histologically proven cases. Medicine (Baltimore). 1995;74:350–358. doi: cholesterol crystals in atheroembolism. Br J Exp Pathol. 1976;57:67–77.
10.1097/00005792-199511000-00005 30. Warren BA, Vales O. Electron microscopy of the sequence of events in the
7. Scolari F, Ravani P. Atheroembolic renal disease. Lancet. 2010;375:1650– atheroembolic occlusion of cerebral arteries in an animal model. Br J Exp
1660. doi: 10.1016/S0140-6736(09)62073-0 Pathol. 1975;56:205–215.
8. Belenfant X, Meyrier A, Jacquot C. Supportive treatment improves survival in 31. Pervaiz MH, Durga S, Janoudi A, Berger K, Abela GS. PET/CTA detec-
multivisceral cholesterol crystal embolism. Am J Kidney Dis. 1999;33:840– tion of muscle inflammation related to cholesterol crystal emboli without
850. doi: 10.1016/s0272-6386(99)70415-4 arterial obstruction. J Nucl Cardiol. 2018;25:433–440. doi: 10.1007/
9. Ghanem F, Vodnala D, K Kalavakunta J, Durga S, Thormeier N, s12350-017-0826-y
Subramaniyam P, Abela S, S Abela G. Cholesterol crystal embolization fol- 32. Lau A, Wang S, Jiang J, Haig A, Pavlosky A, Linkermann A, Zhang ZX,
lowing plaque rupture: a systemic disease with unusual features. J Biomed Jevnikar AM. RIPK3-mediated necroptosis promotes donor kidney
Res. 2017;31:82–94. doi: 10.7555/JBR.31.20160100 inflammatory injury and reduces allograft survival. Am J Transplant.
2013;13:2805–2818. doi: 10.1111/ajt.12447
Downloaded from http://ahajournals.org by on May 8, 2023

10. Lusco MA, Najafian B, Alpers CE, Fogo AB. AJKD atlas of renal pathology:
cholesterol emboli. Am J Kidney Dis. 2016;67:e23–e24. doi: 10.1053/j. 33. Linkermann A, Bräsen JH, Himmerkus N, Liu S, Huber TB, Kunzendorf
ajkd.2016.02.034 U, Krautwald S. Rip1 (receptor-interacting protein kinase 1) mediates
11. Mulay SR, Anders HJ. Crystal nephropathies: mechanisms of crystal- necroptosis and contributes to renal ischemia/reperfusion injury. Kidney Int.
induced kidney injury. Nat Rev Nephrol. 2017;13:226–240. doi: 10.1038/ 2012;81:751–761. doi: 10.1038/ki.2011.450
nrneph.2017.10 34. Meyrier A. Cholesterol crystal embolism: diagnosis and treatment. Kidney Int.
12. Murphy JM, Czabotar PE, Hildebrand JM, Lucet IS, Zhang JG, Alvarez-Diaz 2006;69:1308–1312. doi: 10.1038/sj.ki.5000263
S, Lewis R, Lalaoui N, Metcalf D, Webb AI, et al. The pseudokinase 35. Diethelm A, Vorburger C, Anabitarte M, Haag E. [Cholesterol crystal embo-
MLKL mediates necroptosis via a molecular switch mechanism. Immunity. lisms due to systemic thrombolysis of an acute myocardial infarct]. Schweiz
2013;39:443–453. doi: 10.1016/j.immuni.2013.06.018 Med Wochenschr. 1994;124:1437–1441.
13. Anders HJ. Necroptosis in acute kidney injury. Nephron. 2018;139:342– 36. Dulíček P, Bártová J, Beránek M, Malý J, Pecka M. The purple toe syn-
348. doi: 10.1159/000489940 drome in female with factor V Leiden mutation successfully treated
14. Liu W, Chen B, Wang Y, Meng C, Huang H, Huang XR, Qin J, Mulay SR, with enoxaparin. Clin Appl Thromb Hemost. 2013;19:100–102. doi:
Anders HJ, Qiu A, et al. RGMb protects against acute kidney injury by 10.1177/1076029612440033
inhibiting tubular cell necroptosis via an MLKL-dependent mechanism. 37. Gutiérrez Solís E, Morales E, Rodríguez Jornet A, Andreu FJ, Rivera F,
Proc Natl Acad Sci U S A. 2018;115:E1475–E1484. doi: 10.1073/pnas. Vozmediano C, Gutiérrez E, Igarzábal A, Enguita AB, Praga M. [Atheroem-
1716959115 bolic renal disease: analysis of clinical and therapeutic factors that influence
15. Martin-Sanchez D, Fontecha-Barriuso M, Carrasco S, Sanchez-Niño MD, its progression]. Nefrologia. 2010;30:317–323. doi: 10.3265/Nefrologia.
Mässenhausen AV, Linkermann A, Cannata-Ortiz P, Ruiz-Ortega M, Egido pre2010.Apr.10367
J, Ortiz A, et al. TWEAK and RIPK1 mediate a second wave of cell death 38. Lekeufack JB, Delrée P, Goergen M, Bouazza F, Salmon K, Raynal P, Azagra
during AKI. Proc Natl Acad Sci U S A. 2018;115:4182–4187. doi: 10.1073/ JS, Sellitti E, Dehon P. [Multiple cholesterol emboli syndrome: beneficial
pnas.1716578115 effects of early heparin therapy. A case report]. Ann Cardiol Angeiol (Paris).
16. von Mässenhausen A, Tonnus W, Himmerkus N, Parmentier S, Saleh D, 1999;48:575–578.
Rodriguez D, Ousingsawat J, Ang RL, Weinberg JM, Sanz AB, et al. Phe- 39. Wakabayashi T, Yoshizawa Y, Kawana S. Successful use of heparin and
nytoin inhibits necroptosis. Cell Death Dis. 2018;9:359. doi: 10.1038/ warfarin in the treatment of cholesterol crystal embolization. J Dermatol.
s41419-018-0394-3 2008;35:111–114. doi: 10.1111/j.1346-8138.2008.00425.x
17. Kim HJ, Lee DW, Ravichandran K, O Keys D, Akcay A, Nguyen Q, He Z, Jani 40. Brill A, Fuchs TA, Savchenko AS, Thomas GM, Martinod K, De Meyer SF,
A, Ljubanovic D, Edelstein CL. NLRP3 inflammasome knockout mice are Bhandari AA, Wagner DD. Neutrophil extracellular traps promote deep
protected against ischemic but not cisplatin-induced acute kidney injury. J vein thrombosis in mice. J Thromb Haemost. 2012;10:136–144. doi:
Pharmacol Exp Ther. 2013;346:465–472. doi: 10.1124/jpet.113.205732 10.1111/j.1538-7836.2011.04544.x
18. Laridan E, Martinod K, De Meyer SF. Neutrophil extracellular traps in arte- 41. Leal AC, Mizurini DM, Gomes T, Rochael NC, Saraiva EM, Dias MS, Werneck
rial and venous thrombosis. Semin Thromb Hemost. 2019;45:86–93. doi: CC, Sielski MS, Vicente CP, Monteiro RQ. Tumor-derived exosomes induce
10.1055/s-0038-1677040 the formation of neutrophil extracellular traps: implications for the estab-
19. Nakazawa D, Desai J, Steiger S, Müller S, Devarapu SK, Mulay SR, Iwakura lishment of cancer-associated thrombosis. Sci Rep. 2017;7:6438. doi:
T, Anders HJ. Activated platelets induce MLKL-driven neutrophil necropto- 10.1038/s41598-017-06893-7
sis and release of neutrophil extracellular traps in venous thrombosis. Cell 42. Boudreau LH, Duchez AC, Cloutier N, Soulet D, Martin N, Bollinger J, Paré
Death Discov. 2018;4:6. doi: 10.1038/s41420-018-0073-2 A, Rousseau M, Naika GS, Lévesque T, et al. Platelets release mitochondria

Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625 April 10, 2020   e51


Shi et al Crystal Clots in Cholesterol Embolism

serving as substrate for bactericidal group IIA-secreted phospholipase A2 measurement of renal function in conscious mice. Am J Physiol Renal
to promote inflammation. Blood. 2014;124:2173–2183. doi: 10.1182/ Physiol. 2012;303:F783–F788. doi: 10.1152/ajprenal.00279.2012
Original Research

blood-2014-05-573543 52. Steiger S, Grill JF, Ma Q, Bäuerle T, Jordan J, Smolle M, Böhland C, Lech
43. McArthur K, Whitehead LW, Heddleston JM, Li L, Padman BS, Oorschot V, M, Anders HJ. Anti-transforming growth factor β IgG elicits a dual effect
Geoghegan ND, Chappaz S, Davidson S, San Chin H, et al. Bak/bax macro- on calcium oxalate crystallization and progressive nephrocalcinosis-
pores facilitate mitochondrial herniation and mtdna efflux during apoptosis. related chronic kidney disease. Front Immunol. 2018;9:619. doi: 10.3389/
Science. 2018;359:eaao6047. doi: 10.1126/science.aao6047 fimmu.2018.00619
44. Gadeela N, Rubinstein J, Tamhane U, Huang R, Pathak DR, Hosein HA, Rich 53. Friedemann J, Heinrich R, Shulhevich Y, Raedle M, William-Olsson L, Pill
M, Dhar G, Abela GS. The impact of circulating cholesterol crystals on vaso- J, Schock-Kusch D. Improved kinetic model for the transcutaneous mea-
motor function: implications for no-reflow phenomenon. JACC Cardiovasc surement of glomerular filtration rate in experimental animals. Kidney Int.
Interv. 2011;4:521–529. doi: 10.1016/j.jcin.2011.02.010 2016;90:1377–1385. doi: 10.1016/j.kint.2016.07.024
45. Falasca GF, Ramachandrula A, Kelley KA, O’Connor CR, Reginato AJ. Super- 54. Liu F, Schafer DP, McCullough LD. TTC, fluoro-Jade B and neuN stain-
oxide anion production and phagocytosis of crystals by cultured endothelial ing confirm evolving phases of infarction induced by middle cerebral
cells. Arthritis Rheum. 1993;36:105–116. doi: 10.1002/art.1780360118
artery occlusion. J Neurosci Methods. 2009;179:1–8. doi: 10.1016/j.
46. Gravastrand CS, Steinkjer B, Halvorsen B, Landsem A, Skjelland M,
jneumeth.2008.12.028
Jacobsen EA, Woodruff TM, Lambris JD, Mollnes TE, Brekke OL, et al. Cho-
55. Feulgen R, Rossenbeck H. Mikroskopisch-chemischer nachweis einer
lesterol crystals induce coagulation activation through complement-depen-
nucleinsäure vom typus des thymonucleinsäure und die darauf beruhende
dent expression of monocytic tissue factor. J Immunol. 2019;203:853–863.
elektive färbung von zellkernen in mikroskopischen präparaten. Hoppe-
doi: 10.4049/jimmunol.1900503
Seylers Z. Physiol Chem. 1924;135:203–248
47. Mani AM, Chattopadhyay R, Singh NK, Rao GN. Cholesterol crystals
56. Mulay SR, Thomasova D, Ryu M, Anders HJ. MDM2 (murine double min-
increase vascular permeability by inactivating SHP2 and disrupting adhe-
ute-2) links inflammation and tubular cell healing during acute kidney injury
rens junctions. Free Radic Biol Med. 2018;123:72–84. doi: 10.1016/j.
freeradbiomed.2018.05.068 in mice. Kidney Int. 2012;81:1199–1211. doi: 10.1038/ki.2011.482
48. Nymo S, Niyonzima N, Espevik T, Mollnes TE. Cholesterol crystal-induced 57. Ehling J, Bábíčková J, Gremse F, Klinkhammer BM, Baetke S, Knuechel
endothelial cell activation is complement-dependent and mediated by TNF. R, Kiessling F, Floege J, Lammers T, Boor P. Quantitative micro-computed
Immunobiology. 2014;219:786–792. doi: 10.1016/j.imbio.2014.06.006 tomography imaging of vascular dysfunction in progressive kidney diseases.
49. Shu F, Chen J, Ma X, Fan Y, Yu L, Zheng W, Amrein MW, Xia T, Shi Y. Choles- J Am Soc Nephrol. 2016;27:520–532. doi: 10.1681/ASN.2015020204
terol crystal-mediated inflammation is driven by plasma membrane destabili- 58. Gremse F, Stärk M, Ehling J, Menzel JR, Lammers T, Kiessling F. Imalyt-
zation. Front Immunol. 2018;9:1163. doi: 10.3389/fimmu.2018.01163 ics preclinical: interactive analysis of biomedical volume data. Theranostics.
50. Marschner JA, Schäfer H, Holderied A, Anders HJ. Optimizing mouse 2016;6:328–341. doi: 10.7150/thno.13624
surgery with online rectal temperature monitoring and preoperative 59. Zimmermann S, Zarse K, Schulz TJ, Siems K, Müller-Kuhrt L, Birringer
heat supply. Effects on post-ischemic acute kidney injury. PLoS One. M, Ristow M. A cell-based high-throughput assay system reveals modu-
2016;11:e0149489. doi: 10.1371/journal.pone.0149489 lation of oxidative and nonoxidative glucose metabolism due to com-
51. Schreiber A, Shulhevich Y, Geraci S, Hesser J, Stsepankou D, Neudecker monly used organic solvents. Horm Metab Res. 2008;40:29–37. doi:
S, Koenig S, Heinrich R, Hoecklin F, Pill J, et al. Transcutaneous 10.1055/s-2007-1004542
Downloaded from http://ahajournals.org by on May 8, 2023

e52   April 10, 2020 Circulation Research. 2020;126:e37–e52. DOI: 10.1161/CIRCRESAHA.119.315625

You might also like