Cytokines are hypothesized to play a central role in the pathophysiology of IgG-mediated hemolytic transfusion reactions (HTRs), and deeper understanding is required for improving therapy for these events. After establishing well-defined mouse models of HTRs, we tested whether cytokines were involved. Red blood cells (RBCs) from human glycophorin A transgenic (hGPA-Tg) or wild-type (WT) mice were transfused into non-Tg recipients passively immunized with monoclonal antibodies (Mabs). Only transfusions of incompatible RBCs induced IgG-mediated HTRs, exemplified by rapid clearance and hemoglobinuria. Very high plasma levels of monocyte chemoattractant protein-1 (MCP-1) and interleukin-6 (IL-6), and lower levels of tumor necrosis factor-α (TNF-α), were induced after incompatible transfusion. No significant changes in IL-10, IL-12, or interferon-γ (IFN-γ) levels were observed. The proinflammatory cytokines elaborated in this in vivo mouse model are also implicated in the systemic inflammatory response syndrome (SIRS) and confirm the hypothesis that cytokine storm occurs as a result of HTRs.

Immune-mediated hemolysis is a serious complication of blood transfusion.1  Hemolytic transfusion reactions (HTRs) vary in severity from clinically inapparent to systemic inflammatory response syndrome (SIRS), multiorgan failure, and death.2  A leading hypothesis regarding the pathophysiology of IgG-mediated HTRs, in which complement is thought to play a minor role, implicates “cytokine storm.”3 

Phagocytosis of IgG-coated red blood cells (RBCs) in vitro induces cytokine secretion, which may cause the clinical symptoms of HTRs.3  Human case reports also suggest that cytokine storm in other settings produces SIRS and multiorgan failure.4  In addition, cytokines were implicated in one human IgG-mediated HTR5  and in patients with autoimmune thrombocytopenic purpura (AITP), who develop hemolysis after receiving anti-Rh(D).6 

Although animal HTR models exist,1,7,8  the role of cytokine storm has not been evaluated. We used a murine HTR model involving passive immunization with IgG1 antibodies, because it is well characterized8  and allows for a more controlled experimental design than the use of active immunization. Thus, in our murine HTR models, passive immunization with anti-human glycophorin A (hGPA) monoclonal antibodies (Mabs) led to rapid, dose-dependent clearance of transfused, incompatible hGPA-transgenic (Tg) RBCs.8  IgG1-mediated clearance was markedly impaired in FcγR knockout (KO) mice, but only moderately inhibited in C3 KO mice, suggesting that activating Fcγ receptors are dominant in this process.8  The current study uses this model to test the hypothesis that HTRs lead to cytokine storm.

Mice

Wild-type (WT) C57BL/6, C3 KO, and FVB/NJ mice were purchased from The Jackson Laboratory (Bar Harbor, ME). hGPA-Tg mice were maintained as described.8  Mice were used at 8 to 12 weeks of age.

Procedures were approved by the Institutional Animal Care and Use Committees at Columbia University Medical Center and at Emory University School of Medicine. No human subjects were used in this study.

Antibodies

IgG1 anti-hGPA (6A7 and 10F7)9  and anti-HEL10  Mabs, purified by protein A chromatography (Bio X Cell, West Lebanon, NH), were quality controlled and confirmed to lack lipopolysaccharide (LPS).8  WT C57BL/6 mice were passively immunized by tail-vein injection with phosphate-buffered saline (PBS), 100 μg 6A7, 2 mg 10F7, or 2 mg anti-HEL before transfusion.

Transfusion of fluorescently labeled RBCs

WT FVB/NJ and hGPA-Tg mice were anesthetized and exsanguinated by cardiac puncture. Washed, buffy coat–depleted RBCs were labeled with chloromethylbenzamido 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate (DiO) per manufacturer's instructions (Invitrogen, Carlsbad, CA). Control, WT C57BL/6 RBCs were labeled with 3,3′-dihexadecyloxacarbocyanine perchlorate (DiO), mixed 1:3 with DiI-labeled RBCs, and brought to a 40% hematocrit with LPS-free PBS. Each mouse was transfused with a 500 μL mixture of either (1) DiI-labeled WT FVB/NJ and DiO-labeled WT C57BL/6 RBCs (compatible transfusion), or (2) DiI-labeled hGPA-Tg and DiO-labeled WT C57BL/6 RBCs (incompatible transfusion).

RBC survival

At defined intervals posttransfusion, mice were anesthetized and 25 μL of retroorbital plexus blood was obtained. To determine percent survival of transfused RBCs, the ratio of DiI-labeled to DiO-labeled RBCs in passively immunized mice was compared with the ratio of DiI-labeled to DiO-labeled RBCs in nonimmunized control mice.10  When possible, urine was collected at necropsy at the final, 20 hour time point. Hemoglobinuria was detected using a PowerWave XS (BioTek, Winooski, VT) spectrophotometer.

Cytokine measurements

Cytokines (interleukin [IL]-6, IL-10, monocyte chemoattractant protein-1 [MCP-1], interferon [IFN]-γ, tumor necrosis factor [TNF]-α, and IL12-p70) were quantified using the Cytometric Bead Array Mouse Inflammation Kit (BD Biosciences, San Diego, CA). Plasma, obtained by retroorbital phlebotomy at various time points (2 hours, 7-9 hours, 17-21 hours) after HTR, was analyzed at a 1:2 and/or 1:10 dilution. Flow cytometry data, acquired with a FACSCan flow cytometer (BD Biosciences),11  was analyzed using FlowJo software (TreeStar, Ashland, OR).

Statistical analysis

One-way ANOVA was used to determine significant differences in cytokine levels and RBC survival using Prism (Graphpad Software; San Diego, CA). A P value of less than .05 was considered significant.

Transfused incompatible RBCs are rapidly cleared in passively immunized mice

Transfused, incompatible hGPA-Tg RBCs were cleared within 2 hours by mice passively immunized with anti-hGPA Mabs (Figure 1A,B). In contrast, transfused WT FVB/NJ RBCs circulated normally (Figure 1A,B). Passive immunization with an irrelevant, isotype-matched control Mab did not affect RBC survival (Figure 1C). Hemoglobinuria, detected by a prominent Soret band, was found in 3 of 4 mice receiving incompatible transfusions, but not in any mice receiving compatible RBCs (n = 9; Figure 1D).

Figure 1

IgG-mediated clearance of transfused incompatible hGPA-Tg RBCs. WT C57BL/6 mice were passively immunized with 2 mg 10F7 anti-hGPA IgG1 Mab (A), 100 μg 6A7 anti-hGPA IgG1 Mab (B), or 2 mg anti-HEL IgG1 Mab (C). Mice were then transfused with DiO-labeled WT C57BL/6 RBCs (as an internal negative control) and with either DiI-labeled incompatible hGPA-Tg RBCs (△) or DiI-labeled compatible WT FVB/NJ RBCs (▲). RBC survival, as percentage of PBS control (ie, nonimmunized) mice, was quantified by flow cytometry at the indicated time points. Five mice were evaluated at each time point; the means plus or minus 1 SD are provided. The results shown are from one representative experiment (from a total of 4 replicates). *P < .05. Urine was obtained at the 20-hour time point from 4 mice receiving incompatible hGPA-Tg RBC transfusions (●) and 9 mice receiving compatible WT FVB/NJ RBC transfusions (x). Representative spectra from one mouse from each group are shown (D).

Figure 1

IgG-mediated clearance of transfused incompatible hGPA-Tg RBCs. WT C57BL/6 mice were passively immunized with 2 mg 10F7 anti-hGPA IgG1 Mab (A), 100 μg 6A7 anti-hGPA IgG1 Mab (B), or 2 mg anti-HEL IgG1 Mab (C). Mice were then transfused with DiO-labeled WT C57BL/6 RBCs (as an internal negative control) and with either DiI-labeled incompatible hGPA-Tg RBCs (△) or DiI-labeled compatible WT FVB/NJ RBCs (▲). RBC survival, as percentage of PBS control (ie, nonimmunized) mice, was quantified by flow cytometry at the indicated time points. Five mice were evaluated at each time point; the means plus or minus 1 SD are provided. The results shown are from one representative experiment (from a total of 4 replicates). *P < .05. Urine was obtained at the 20-hour time point from 4 mice receiving incompatible hGPA-Tg RBC transfusions (●) and 9 mice receiving compatible WT FVB/NJ RBC transfusions (x). Representative spectra from one mouse from each group are shown (D).

Close modal

Approximately 20-fold more 10F7 than 6A7 was required to induce similar degrees of clearance in vivo (Figure 1) and phagocytosis by mouse macrophages12  in vitro (E.A.H., S.A.S., S.L.S. unpublished data, November 2007). This may result from differences in specificity, affinity, or effector function. For example, although both are IgG1 Mabs, their glycosylation differs (E.A.H., D.A.S., S.L.S., unpublished data, June 2007), and Fc glycosylation modulates effector function.13  Future studies will evaluate the effect of Fc glycosylation on IgG-mediated HTRs.

Some mouse and human IgG antibodies fix complement via the classical pathway, thereby causing intravascular hemolysis, hemoglobinuria, and renal damage.14,15  For example, we previously showed that clearance of incompatible RBCs by 10F7 was moderately impaired in C3 KO mice.8  In the current study, we evaluated whether complement was required for cytokine storm in IgG-induced HTRs (see below; E.A.H., J.C.Z., S.L.S., unpublished data, April 2008). Interestingly, hemoglobinuria was seen in some C3 KO mice after clearance of incompatible RBCs by 10F7 or 6A7 (not shown). Finding IgG-induced hemoglobinuria in C3-deficient mice is surprising, but may result from the ability of some anti-hGPA antibodies to destabilize RBC phospholipid bilayers, thereby inducing hemolysis.16  In addition, the ability of thrombin to cleave C5 in C3 KO mice may provide another explanation for this phenomenon.17 

IgG-mediated HTRs induce cytokine storm

Plasma levels of 6 classical pro- and antiinflammatory cytokines, which prior in vitro studies2  suggested were relevant for IgG-mediated HTRs, were measured at various times after initiation of HTRs. Significantly increased levels of the chemokine MCP-1 and the proinflammatory cytokines IL-6 and TNF-α were detected at 2 hours after incompatible transfusion (Figure 2A-C); these returned to baseline by 7 to 9 hours after transfusion, at the latest. IL-10, IL-12, and IFN-γ levels remained low at all tested time points (Figure 2D-F). In addition, 6A7 and 10F7 induced similar cytokine profiles in C3 KO and WT mice (E.A.H., J.C.Z., S.L.S., unpublished data, April 2008). Therefore, cleavage of the C3 component of complement is not required for cytokine storm. Future studies will determine in detail what role, if any, the complement system plays in producing cytokine storm in this setting.

Figure 2

Plasma cytokine concentrations in C57BL/6 mice at 2, 7, and 20 hours after transfusion. WT C57BL/6 mice were passively immunized with 2 mg 10F7 anti-hGPA IgG1 Mab, 100 μg 6A7 anti-hGPA IgG1 Mab, or 2 mg anti-HEL (irrelevant, isotype-matched control) IgG1 Mab. Mice were transfused with DiO-labeled WT C57BL/6 RBCs (as a negative, internal control) and with either DiI-labeled incompatible hGPA-Tg RBCs (data shown) or compatible WT FVB/NJ RBCs (data not shown because all of these results are very similar to the anti-HEL controls). IL-6 (A), TNF-α (B), MCP-1 (C), IFN-γ (D), IL-10 (E), and IL-12p70 (F) concentrations were measured in plasma, as described in “Cytokine measurements” (note that the scale on the y-axis varies from graph to graph and the absence of bars represents results below the limit of detection of the assay). Five mice were evaluated at each time point and the mean plus or minus 1 SD is provided. Data shown are from 1 representative experiment (from a total of 4 replicates). *P < .05 compared with the anti-HEL controls.

Figure 2

Plasma cytokine concentrations in C57BL/6 mice at 2, 7, and 20 hours after transfusion. WT C57BL/6 mice were passively immunized with 2 mg 10F7 anti-hGPA IgG1 Mab, 100 μg 6A7 anti-hGPA IgG1 Mab, or 2 mg anti-HEL (irrelevant, isotype-matched control) IgG1 Mab. Mice were transfused with DiO-labeled WT C57BL/6 RBCs (as a negative, internal control) and with either DiI-labeled incompatible hGPA-Tg RBCs (data shown) or compatible WT FVB/NJ RBCs (data not shown because all of these results are very similar to the anti-HEL controls). IL-6 (A), TNF-α (B), MCP-1 (C), IFN-γ (D), IL-10 (E), and IL-12p70 (F) concentrations were measured in plasma, as described in “Cytokine measurements” (note that the scale on the y-axis varies from graph to graph and the absence of bars represents results below the limit of detection of the assay). Five mice were evaluated at each time point and the mean plus or minus 1 SD is provided. Data shown are from 1 representative experiment (from a total of 4 replicates). *P < .05 compared with the anti-HEL controls.

Close modal

Qualitatively consistent with these novel in vivo results, previous in vitro IgG-mediated HTR models identified high levels of MCP-1 and low levels of IL-6 and TNF-α.18  MCP-1, important for leukocyte recruitment, activation, and phagocytosis, is secreted by macrophages, lymphocytes, platelets, and endothelial cells.19  IL-6 and TNF-α induce fever, the acute-phase response, and activation of innate and adaptive immunity.3  Both were elevated in an isolated human HTR case,5  and IL-6 was implicated in human febrile nonhemolytic transfusion reactions.20  AITP patients receiving anti-Rh(D) can have elevated IL-6, TNF-α, IL-10, and MCP-1 by 2 hours after treatment.6 

In mouse models of IgG-mediated autoimmune hemolytic anemia, RBCs are predominantly cleared by Kupffer cells,21  which also release MCP-1 and IL-6 in trauma-hemorrhage models.19  Thus, Kupffer cells probably clear RBCs and produce cytokines in our model; however, other sources are possible and further studies are necessary.

In general, the array of inflammatory cytokines expressed varies with the nature of the inflammatory insult.22  For example, in mice, all 6 cytokines evaluated in the current study are elevated within 2 hours of LPS infusion.23  In addition, symptom severity correlates with cytokine levels.24  Therefore, ongoing studies, which will provide a detailed understanding of the profile and amplitude of cytokines induced by HTRs, will lead to a deeper understanding of the pathophysiology of IgG-mediated HTRs, thereby leading to the rational design of novel therapeutic interventions.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank Traci Chadwick for outstanding technical assistance.

This work was supported in part by a grant from the National Institutes of Health (R21 HL987906; S.L.S. and J.C.Z.).

National Institutes of Health

Contribution: All authors participated in designing and performing the research; E.A.H., S.A.S., J.C.Z., and S.L.S. controlled and analyzed the data; J.J. performed the statistical analysis; E.A.H. and S.L.S. wrote the paper; and all authors critiqued various drafts and checked the final version of the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Steven L. Spitalnik, Department of Pathology and Cell Biology, 630 West 168th St, College of Physicians and Surgeons of Columbia University, New York, NY 10023; e-mail: ss2479@columbia.edu.

1
Ness
 
PM
Shirey
 
RS
Weinstein
 
MH
King
 
KE
An animal model for delayed hemolytic transfusion reactions.
Transfus Med Rev
2001
, vol. 
15
 (pg. 
305
-
317
)
2
Davenport
 
RD
Pathophysiology of hemolytic transfusion reactions.
Semin Hematol
2005
, vol. 
42
 (pg. 
165
-
168
)
3
Davenport
 
RD
Burdick
 
M
Moore
 
SA
Kunkel
 
SL
Cytokine production in IgG-mediated red cell incompatibility.
Transfusion
1993
, vol. 
33
 (pg. 
19
-
24
)
4
Suntharalingam
 
G
Perry
 
MR
Ward
 
S
, et al. 
Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412.
N Engl J Med
2006
, vol. 
355
 (pg. 
1018
-
1028
)
5
von Zabern
 
I
Ehlers
 
M
Grunwald
 
U
Mauermann
 
K
Greinacher
 
A
Release of mediators of systemic inflammatory response syndrome in the course of a severe delayed hemolytic transfusion reaction caused by anti-D.
Transfusion
1998
, vol. 
38
 (pg. 
459
-
468
)
6
Cooper
 
N
Heddle
 
NM
Haas
 
M
, et al. 
Intravenous (IV) anti-D and IV immunoglobulin achieve acute platelet increases by different mechanisms: modulation of cytokine and platelet responses to IV anti-D by FcgammaRIIa and FcgammaRIIIa polymorphisms.
Br J Haematol
2004
, vol. 
124
 (pg. 
511
-
518
)
7
Campbell-Lee
 
SA
Liu
 
J
Velliquette
 
RW
, et al. 
The production of red blood cell alloantibodies in mice transfused with blood from transgenic Fyb-expressing mice.
Transfusion
2006
, vol. 
46
 (pg. 
1682
-
1688
)
8
Schirmer
 
DA
Song
 
SC
Baliff
 
JP
, et al. 
Mouse models of IgG- and IgM-mediated hemolysis.
Blood
2007
, vol. 
109
 (pg. 
3099
-
3107
)
9
Bigbee
 
WL
Langlois
 
RG
Vanderlaan
 
M
Jensen
 
RH
Binding specificities of eight monoclonal antibodies to human glycophorin A–studies with McM, and MkEn(UK) variant human erythrocytes and M- and MNV-type chimpanzee erythrocytes.
J Immunol
1984
, vol. 
133
 (pg. 
3149
-
3155
)
10
Zimring
 
JC
Cadwell
 
CM
Chadwick
 
TE
, et al. 
Nonhemolytic antigen loss from red blood cells requires cooperative binding of multiple antibodies recognizing different epitopes.
Blood
2007
, vol. 
110
 (pg. 
2201
-
2208
)
11
Chen
 
R
Lowe
 
L
Wilson
 
JD
, et al. 
Simultaneous quantification of six human cytokines in a single sample using microparticle-based flow cytometric technology.
Clin Chem
1999
, vol. 
45
 (pg. 
1693
-
1694
)
12
Raschke
 
WC
Baird
 
S
Ralph
 
P
Nakoinz
 
I
Functional macrophage cell lines transformed by Abelson leukemia virus.
Cell
1978
, vol. 
15
 (pg. 
261
-
267
)
13
Kaneko
 
Y
Nimmerjahn
 
F
Ravetch
 
JV
Anti-inflammatory activity of immunoglobulin G resulting from Fc sialylation.
Science
2006
, vol. 
313
 (pg. 
670
-
673
)
14
Salama
 
A
Mueller-Eckhardt
 
C
Delayed hemolytic transfusion reactions. Evidence for complement activation involving allogeneic and autologous red cells.
Transfusion
1984
, vol. 
24
 (pg. 
188
-
193
)
15
Azeredo da Silveira
 
S
Kikuchi
 
S
Fossati-Jimack
 
L
, et al. 
Complement activation selectively potentiates the pathogenicity of the IgG2b and IgG3 isotypes of a high affinity anti-erythrocyte autoantibody.
J Exp Med
2002
, vol. 
195
 (pg. 
665
-
672
)
16
Brain
 
MC
Prevost
 
JM
Pihl
 
CE
Brown
 
CB
Glycophorin A-mediated haemolysis of normal human erythrocytes: evidence for antigen aggregation in the pathogenesis of immune haemolysis.
Br J Haematol
2002
, vol. 
118
 (pg. 
899
-
908
)
17
Huber-Lang
 
M
Sarma
 
JV
Zetoune
 
FS
, et al. 
Generation of C5a in the absence of C3: a new complement activation pathway.
Nat Med
2006
, vol. 
12
 (pg. 
682
-
687
)
18
Davenport
 
RD
Cytokines as intercellular signals in hemolytic transfusion reactions.
Biol Signals
1996
, vol. 
5
 (pg. 
240
-
245
)
19
Hildebrand
 
F
Hubbard
 
WJ
Choudhry
 
MA
, et al. 
Kupffer cells and their mediators: the culprits in producing distant organ damage after trauma-hemorrhage.
Am J Pathol
2006
, vol. 
169
 (pg. 
784
-
794
)
20
Sacher
 
RA
Boyle
 
L
Freter
 
CE
High circulating interleukin 6 levels associated with acute transfusion reaction: cause or effect?
Transfusion
1993
, vol. 
33
 (pg. 
962
-
963
)
21
Fossati-Jimack
 
L
Ioan-Facsinay
 
A
Reininger
 
L
, et al. 
Markedly different pathogenicity of four immunoglobulin G isotype-switch variants of an antierythrocyte autoantibody is based on their capacity to interact in vivo with the low-affinity Fcgamma receptor III.
J Exp Med
2000
, vol. 
191
 (pg. 
1293
-
1302
)
22
Clark
 
IA
The advent of the cytokine storm.
Immunol Cell Biol
2007
, vol. 
85
 (pg. 
271
-
273
)
23
Gonzalez-Rey
 
E
Chorny
 
A
Varela
 
N
Robledo
 
G
Delgado
 
M
Urocortin and adrenomedullin prevent lethal endotoxemia by down-regulating the inflammatory response.
Am J Pathol
2006
, vol. 
168
 (pg. 
1921
-
1930
)
24
Netea
 
MG
van der Meer
 
JW
van Deuren
 
M
Kullberg
 
BJ
Proinflammatory cytokines and sepsis syndrome: not enough, or too much of a good thing?
Trends Immunol
2003
, vol. 
24
 (pg. 
254
-
258
)
Sign in via your Institution