ArticlePDF Available

Synergistic Inflammation Is Induced by Blood Degradation Products with Microbial Toll-Like Receptor Agonists and Is Blocked by Hemopexin

Authors:

Abstract and Figures

Detection of microbial components by immune cells via Toll-like receptors (TLRs) with subsequent induction of inflammation is essential for host defense. However, an overactive immune response can cause tissue damage and sepsis. The endogenous molecule hemoglobin and its derivative heme are often released into tissue compartments where there is infection in the presence of degrading blood. We found that hemoglobin synergizes with multiple TLR agonists to induce high levels of tumor necrosis factor and interleukin-6 from macrophages and that this synergy is independent of TLR4 and MyD88. In contrast, heme synergized with some but not all TLR agonists studied. Furthermore, the synergy of both hemoglobin and heme with lipopolysaccharide was suppressed by hemopexin, a plasma heme-binding protein. These studies suggest that hemoglobin and heme may substantially contribute to microbe-induced inflammation when bacterial or viral infection coexists with blood degradation and that hemopexin may play a role in controlling inflammation in such settings.
Content may be subject to copyright.
Synergistic Inflammation Is Induced by Blood Degradation
Products with Microbial Toll-Like Receptor Agonists and Is
Blocked by Hemopexin
Tian Lin1,a, Young Ho Kwak1,a, Fatima Sammy1, Ping He1, Sujatha Thundivalappil1,
Guangjie Sun1, Wei Chao2, and H. Shaw Warren1
1 Infectious Disease Unit, Massachusetts General Hospital, Boston
2 Department of Anesthesia and Critical Care, Massachusetts General Hospital, Boston
Abstract
Detection of microbial components by immune cells via Toll-like receptors (TLRs) with
subsequent induction of inflammation is essential for host defense. However, an overactive
immune response can cause tissue damage and sepsis. The endogenous molecule hemoglobin and
its derivative heme are often released into tissue compartments where there is infection in the
presence of degrading blood. We found that hemoglobin synergizes with multiple TLR agonists to
induce high levels of tumor necrosis factor and interleukin-6 from macrophages and that this
synergy is independent of TLR4 and MyD88. In contrast, heme synergized with some but not all
TLR agonists studied. Furthermore, the synergy of both hemoglobin and heme with lipo-
polysaccharide was suppressed by hemopexin, a plasma heme-binding protein. These studies
suggest that hemoglobin and heme may substantially contribute to microbe-induced inflammation
when bacterial or viral infection coexists with blood degradation and that hemopexin may play a
role in controlling inflammation in such settings.
Much of the pathophysiology that occurs early and during microbial infection is believed to
be due to the induction of inflammation in tissues that has evolved as an essential part of the
defense against early microbial challenge. Integral to this concept is the early identification
of microbes in tissues by specialized cells, such as macrophages, with the production of
secondary mediators, such as cytokines, that amplify the signal and communicate with other
local and distant tissues. It is now appreciated that certain molecules on microorganisms
known as microbial-associated molecular pattern molecules (MAMPs) interact with a
limited number of pattern recognition receptors called Toll-like receptors (TLRs), to initiate
a cascade of events that ultimately result in a signal being transmitted to the nucleus to
produce cytokines. Each TLR has receptor-specific ligands, such as lipopolysaccharide
(LPS), which signals through TLR4; lipoteichoic acid and peptidoglycan, which signal
through TLR2; viral nucleic acid structures, such as double-stranded RNA poly I: C, which
signals through TLR3; the guanosine analogue loxoribine, which signals through TLR7; and
bacterial DNA (CpG), which signals through TLR9 [1,2].
In some clinical situations, a hyperactive immune response can cause tissue damage. Sepsis
syndrome, which is defined by certain parameters of systemic inflammation in the setting of
Reprints or correspondence: H. Shaw Warren, Infectious Disease Unit, Massachusetts General Hospital East, 149 13th St, 5th Fl,
Charlestown, MA 02129 (swarren1@partners.org).
aThese two authors contributed equally to this work.
Potential conflicts of interest: in accordance with institutional policy, H.S.W. has reported the use of hemopexin as a potential anti-
inflammatory agent, and an application for patent protection has been filed. All other authors: no conflicts.
NIH Public Access
Author Manuscript
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
Published in final edited form as:
J Infect Dis
. 2010 August 15; 202(4): 624–632. doi:10.1086/654929.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
infection [3], is characterized at early stages by high levels of proinflammatory cytokines
[4]. Although the precise mechanisms underlying sepsis syndrome are not well understood,
it is widely believed that excessive production of cytokines may be a driving force, a
situation sometimes referred to as a “cytokine storm.” A better understanding of the
mechanisms responsible for the liberation of these cytokines may permit the development of
effective control strategies.
Exogenous microbial TLR ligands synergize with one another to activate signaling pathways
with subsequent induction of proinflammatory cytokines from macrophages and other
immune cells [5–10]. Data from our laboratory suggest that the outcome of stimulation with
different microbial TLR ligands is dependent on differential engagement of MyD88-
dependent and MyD88-independent pathways [11]. Recent studies suggest that endogenous
host molecules also can act as TLR ligands and that molecules released or induced during
tissue damage may contribute to the induction of inflammatory cytokines in sepsis [4]. For
example, such oxidants as hydrogen peroxide [12], heat-shock proteins (HSPs; ie, HSP-60,
HSP-70, and Gp-96), and self-messenger RNA have been proposed to synergize with
exogenous TLR agonists [13].
Visible or microscopic blood is often present in tissues where there is infection and necrosis,
so that hemoglobin and microorganisms coexist in infected microenvironments. This
situation is particularly common when there is invasive bacterial or viral infection with
tissue necrosis; after trauma, burns, or surgery; or in any infection where there is breach of
capillaries. Older studies revealed that blood and hemoglobin enhanced growth of bacteria
by providing heme as a nutrient source [14]. However, hemoglobin is also known to
synergize with LPS to augment macrophage induction of tumor necrosis factor (TNF) [15–
18]. It has been proposed that hemoglobin preparations increase the biological activity of
LPS through physically interacting with LPS [19,20]. Many of the studies involving the
interactions of hemoglobin and LPS have focused on the development of artificially cross-
linked hemoglobin for use as a cell-free blood transfusion substitute, where such an
interaction has major potential implications [15,16,21,22].
Given the ubiquity of blood in infected tissues and the broad array of bacterial and viral
infections in which blood might play a role, we studied the activation of macrophages in the
presence of hemoglobin and multiple different TLR ligand agonists (ie, ligands for TLR2,
TLR3, TLR4, TLR7, and TLR9). We observed extensive synergy with all of these TLR
agonists. We found that this synergy is not TLR4 dependent and not MyD88 dependent and
that the degraded hemoglobin product, hemin, synergizes with some (but not all) TLR
agonists. Finally, we observed that hemopexin (Hx), a plasma protein that binds heme with
an extremely high affinity, blocks the synergy of both hemoglobin and hemin with LPS,
raising the possibility that Hx may be involved in local regulation of this synergy. Our
findings also suggest that exogenously administered Hx might be a candidate for treating
inflammation induced by microorganisms in tissues that contain blood breakdown products
through a novel mechanism of blocking this synergistic activation of inflammation.
MATERIALS AND METHODS
Materials
The following TLR agonists were purchased: smooth LPS from Escherichia coli O55:B5
(List Biologicals), Pam3Cys (EMC Microcollections), poly I:C, loxoribine, and CpG DNA
(Invivogen). All these TLR agonists were dissolved in pyrogen-free H2O and saved as
aliquots at 80°C. E. coli O4 was the kind gift of Alan Cross (University of Maryland), and
it was heat killed and washed before use, as described elsewhere [23]. Heat-killed
Staphylococcus aureus was purchased from Invivogen. Hemin chloride was purchased from
Lin et al. Page 2
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Frontier Scientific. Hemin solutions were made immediately before use in the dark, as
described elsewhere [24]. In brief, the powder was dissolved in pyrogen-free 0.1 N NaOH at
10 mg/mL and was further diluted in serum-free medium as desired; these steps were
followed by filtration through 0.22-μm Millipore membranes. C57BL/6, C3H/HeN, and
C3H/HeJ mice were obtained from Charles River Laboratories. MyD88 knockout
(MyD88/) mice were generated by Kawai and colleagues [25] and had been backcrossed
>10 generations into the C57BL/6J strain.
Purification of mouse hemoglobin
Hemoglobin was purified as previously described elsewhere [26], with modifications, under
pyrogen-free conditions. In brief, mouse blood was collected from C57BL/6 mice by means
of cardiac puncture, and it was washed with an equal weight of isotonic saline solution
(0.9% NaCl, wt/vol) three times by centrifugation at 1000 g, to remove serum proteins.
Equal volumes of saline were added to the pellet containing red blood cells, and this solution
was sonicated for 5 × 10 s at 40% amplitude with 1-min laps between pulses in a Branson
450 sonicator from Branson Ultrasonics. The hemoglobin solution was diluted with an equal
volume of saline and subjected to a second centrifugation at 2000 g for 1 h. The resulting
hemoglobin solution, removed from the center layer, was filtered through 0.22-μm Millipore
membranes and saved at 20°C in the dark. The concentration of purified mouse
hemoglobin was measured using Micro-BCA. The purity of the hemoglobin was confirmed
to be >99%, by means of nondenaturing polyacrylamide gel electrophoresis and high-
pressure liquid chromatography.
Purification of Hx from mouse or human serum
Mouse serum Hx (mHx) or human serum Hx (hHx) was purified using heme affinity
chromatography essentially as we have described elsewhere [27]. In brief, after filtration
through 0.22- μm Millipore membranes, the abundant albumin in the serum was precipitated
and removed by use of cold 1.68% rivanol solution (pH 8.0). The sample obtained after
rivanol precipitation was dialyzed against pyrogen-free phosphate-buffered saline. Protease
inhibitors (0.5 mmol/L 4-[2-aminoethyl] ben-zenesulfonyl fluoride, 10 μmol/L E-64, 2 μg/
mL aprotinin, and 1 μmol/L pepstatin A) were added, and they interacted with the dialyzed
sample obtained after rivanol precipitation for 15 min by gentle agitation at 4°C. The
mixture was applied to 6 mL of heminagarose column (Sigma) 3 times, followed by
extensive washing with 1200 mL of phosphate-buffered saline containing 0.5 mol/L NaCl
overnight at 4°C to remove unbound proteins. Hx bound to the column was eluted by 0.2
mol/L citric acid (pH 2.0), followed by immediate neutralization with 10 mol/L NaOH.
Proteins in the buffer were exchanged, concentrated in PBS at 4°C by use of Centriprep
YM- 30 (Millipore), and saved in aliquots at 80°C.
Limulus amebocyte lysate assay
The limulus amebocyte lysate assay was performed as previously described elsewhere [28].
Preparation of macrophages
Bone marrow–derived macrophages (BMDMs) were prepared from mice, as described
elsewhere [29], with minor modifications [11,27]. BMDMs were seeded at 1.28 × 105 cells/
well in 96-well tissue culture plates and were allowed to adhere overnight before use in
assays.
Macrophage assays and cytokine production
BMDMs were washed 3 times in serum-free medium, followed by incubation overnight with
different TLR agonists with or without hemoglobin or hemin chloride at indicated
Lin et al. Page 3
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
concentrations. Purified mHx was added to the culture in some experiments, as noted.
Levels of TNF and interleukin (IL)–6 in the supernatants were quantitated by enzyme-linked
immunosorbent assay (ELISA; R & D Systems), in accordance with the manufacturer’s
instructions.
Human whole-blood assay
Whole-blood stimulations were performed as previously described elsewhere [30].
Heparinized whole blood, collected aseptically from healthy human volunteers, was diluted
1:4 in pyrogen-free RPMI 1640 (Cellgro; Mediatech) and placed in a 96-well plate at 100
μL/well. The diluted blood was incubated with desired concentrations of stimuli at 37°C,
followed by centrifugation at 500 g for 5 min. The supernatants were saved for
quantification of cytokines by ELISA.
Statistics
Except where indicated, representative data from at least three experiments are presented in
the figures. Data are expressed as means, and error bars denote standard errors. The data
were analyzed using Prism 5 software (GraphPad). One-way analysis of variance followed
by Dunnett’s post hoc test was used to assess cytokine levels produced by macrophages
treated with different TLR agonists in the absence or presence of hemin or hemoglobin at
desired concentrations. Student’s t test was used to compare the cytokine levels produced by
macrophages from C3H/HeN with C3H/HeJ or C57BL/6 with MyD88/ mice in the
presence of TLR agonists and hemin or hemoglobin at desired concentrations. Two-way
analysis of variance followed by Bonferroni’s post hoc test was used to assess cytokine
levels from macrophages treated with LPS with or without hemin or hemoglobin at different
concentrations in the presence of Hx or phosphate-buffered saline. P <.05 was considered to
denote statistical significance.
RESULTS
Strong synergy of hemoglobin with TLR2, TLR3, TLR4, TLR7, and TLR9 agonists and with
bacteria to induce TNF and IL-6 from macrophages
Differing concentrations of hemoglobin were incubated with a predetermined optimized
concentration of each TLR agonist in cell culture with BMDMs, as described in Materials
and Methods. Levels of the proinflammatory cytokines TNF and IL-6 induced in the
supernatant were measured by ELISA. Hemoglobin significantly enhanced the induction of
TNF and IL-6 from BMDMs by all TLR agonist ligands studied (LPS, Pam3Cys, Poly I:C,
loxoribine, and CpG) in a dose-dependent manner (Figure 1). Concentrations of hemoglobin
as low as 30 μg/mL led to synergistic induction of the cytokines with LPS and Pam3Cys
(Figure 1A–D), whereas higher concentrations (~300 μg/mL) of hemoglobin were required
for loxoribine (Figure 1G, H) and CpG DNA (Figure 1I and 1J). Similar results were
obtained with different concentrations of the TLR agonists (data not shown) as well as with
killed E. coli and S. aureus (Figure 2A, 2B, 2E, and 2F). Synergy was observed as early as 3
h after the cells were stimulated (Figure 2C, 2D, 2G, and 2H).
TLR4- and MyD88-independent synergistic induction of proinflammatory cytokines by
hemoglobin and various TLR agonists
Because heme has been described as a TLR4 ligand agonist [24], we next addressed the
question of whether hemoglobin acts through TLR4-signaling pathways to induce the
synergy that we observed. For these experiments, the synergistic induction of cytokines
induced in BMDMs from C3H/HeJ mice, which are deficient in TLR4, was compared with
induction of cytokines from control BMDMs from C3H/HeN mice, by use of the same TLR
Lin et al. Page 4
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
agonists shown in Figure 1. As expected, neither LPS nor LPS with hemoglobin induced
TNF and IL-6 from C3H/HeJ macrophages (data not shown). The synergy for non-LPS
ligands with hemoglobin was present despite the deficiency of TLR4 (Figure 3), suggesting
that heme ligation of TLR4 [24] was not a prominent mechanism in the synergy. To be able
to compare between cell preparations from the different strains, these results are presented
as the percentage change.
We previously noted that the synergy between different microbial TLR agonists is altered
depending on whether the agonists function through the MyD88-dependent signaling
pathway [11]. MyD88 is an essential adaptor molecule in the signaling pathway of many
TLRs, with the exception that TLR3 is exclusively and TLR4 is partially, but not
predominantly, MyD88 independent [11]. Accordingly, we assessed the synergy of
hemoglobin with ligands of TLR3 and TLR4 in BMDMs from MyD88/ mice. As shown
in Figure 4, hemoglobin induced a similar percentage increase in TNF from both wild-type
(C57BL/6) and MyD88-deficient BMDMs, when stimulated with TLR4 agonist LPS and
TLR3 agonist poly I:C. There was no significant IL-6 produced from MyD88-deficient
BMDMs (not shown). As expected, other TLR agonists (ie, Pam3Cys, loxoribine, and CpG
DNA) did not induce TNF and IL-6 from MyD88-deficient BMDMs, with or without
hemoglobin (data not shown).
Synergistic induction of proinflammatory cytokines from macrophages with hemin and
agonists of TLR3, TLR4, and TLR7—but not with TLR2 and TLR9—and the role of TLR4
and MyD88
Hemoglobin consists of 2 moieties: hemin and globin. It has been hypothesized but not
directly shown that hemin is the active moiety that synergizes with LPS [31]. To directly
study this question, we evaluated the synergy of hemin chloride in the presence and absence
of different TLR agonists. As previously reported, we found that hemin alone weakly but
significantly enhanced production of TNF (data not shown) [24]. Hemin synergized with
LPS (Figure 5A), poly I:C (Figure 5B), and loxoribine (Figure 5C). We also found that the
synergy of hemin with poly I:C and loxoribine was not TLR4 dependent (Figure 5B and 5C)
and that synergy with LPS was partially but not completely MyD88 dependent (Figure 5D
and 5E). Of note, we did not find any hemin synergy with Pam3Cys and CpG DNA,
although a large range of hemin chloride concentrations (0.01 to ~30 μmol/L) was tested
(data not shown).
Suppressive effect of Hx on the synergistic induction of pro-inflammatory cytokines from
macrophages by hemin and hemoglobin with LPS
Hx is the major heme scavenger in plasma and has been described to bind free heme in a 1:1
ratio with remarkably high affinity [32,33]. In addition, Hx has some immunomodulatory
activities in that it modestly down-regulates proinflammatory cytokines from macrophages
[27] and functions as an anti-inflammatory component of serum high-density lipoprotein in
atherosclerosis [34]. It was therefore of interest to assess whether Hx would alter the
induction of pro-inflammatory cytokines that were synergistically induced by hemin with
LPS. We found that Hx abrogated the synergistic induction of TNF (Figure 6A). Similar
results were obtained with IL-6 (data not shown). Unexpectedly, Hx also significantly
down-regulated the synergistic induction of TNF by hemoglobin with LPS (Figure 6B).
Similar results were obtained with IL-6 (data not shown). As previously described elsewhere
[27], in both situations, Hx decreased LPS-induced TNF when studied in the absence of
hemin or hemoglobin (as designated by the zero point in Figures 6A and 6B).
Lin et al. Page 5
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Suppressive effect of the induction of proinflammatory cytokines by LPS and E. coli in
whole human blood
A human whole-blood stimulation assay has been proposed as a more physiological system
in which to assess anti-inflammatory drugs [35]. We found that induction of TNF by LPS
and E. coli was significantly reduced by the addition of mHx and hHx in this system (Figure
6C and 6D).
DISCUSSION
Inflammation induced by microbes through activation of TLRs is believed to play a critical
role in host defense; however, in some circumstances, this same inflammation is also
believed to cause sepsis and tissue injury. We previously studied the synergy between
different exogenous microbial TLR agonists, in an effort to dissect the initiation process of
this inflammation [11]. In the present study, we report that multiple TLR agonists synergize
remarkably with 2 endogenous molecules, hemoglobin and its derivative heme. This finding
likely has broad importance in the clinic, because microscopic blood and blood degradation
products are present in many infected local microenvironments. Furthermore, we found that
this synergy is blocked by Hx, suggesting that Hx may be important in controlling the
inflammation in which even small amounts of degraded blood are present.
Older studies revealed that blood and hemoglobin enhance growth of bacteria by providing a
source of heme [14]. Our findings extend prior available information related to the induction
of cytokines by microbial cell walls in the setting of blood. Much of this information comes
from studies to evaluate the safety of using soluble cross-linked hemoglobin preparations as
a potential substitute for red blood cells, although enhancement of LPS-induced
inflammation by native hemoglobin has also been reported in multiple previous studies
[16,18,31]. In addition to the synergy with LPS, hemoglobin has been reported to increase
IL-6, when present in combination with lipoteichoic acid [36], and several cytokines, when
present in combination with the cell wall of Streptococcus suis [37]. To our knowledge, this
is the first study to systematically evaluate the synergy of native hemoglobin on
inflammation induced by multiple TLR agonists, including ligands for TLR3, TLR7, and
TLR9, which are involved in inducing inflammation after viral infections [38,39], to report
synergy of TLR agonists with heme and to report that the synergy is suppressed with Hx.
The mechanisms responsible for the synergy of hemoglobin with different TLR agonists
remain unclear. It has been proposed that one mechanism for hemoglobin synergy with LPS
is through direct binding of hemoglobin to LPS leading to better presentation of LPS to
TLR4 or is by inducing the conformational changes in the lipid A moiety of LPS [19,20].
Other studies suggest that hemoglobin may produce cell hypersensitivity to LPS by
increasing the intracellular load of heme iron, which catalyze cellular redox changes and
oxidant damage without direct interacting with LPS [40]. In a separate study, it was reported
that hemoglobin synergy with lipotechoic acid was partially dependent on TLR4 [36].
Although we cannot exclude the possibility that each of these different TLR agonist
molecules bound to hemoglobin or heme and that presentation was enhanced to the TLR,
this seems unlikely given their markedly different molecular properties. We also found that
TLR4 is not required for hemoglobin synergy with the synthetic TLR2 agonist Pam3Cys as
well as with TLR 3, TLR7, and TLR9 agonists and that MyD88 signaling is not required for
synergy with LPS and poly I:C, which activate cells via partial and exclusive MyD88-
independent pathways, respectively. The reason for the partial dependence on MyD88 for
the heme synergy with LPS is unclear. Taken together, these findings suggest that the role of
hemoglobin itself in the synergy may be independent of TLR activation. One possible
mechanism may be through quenching of the anti-inflammatory action of nitric oxide [41].
Lin et al. Page 6
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Blood and/or blood degradation products in infected body fluids are common. Sites at which
this occurs include but are not limited to pleural infection, peritoneal infection, central
nervous system infection, surgical wounds, burn wounds, trauma wounds, pancreatitis, and
necrotizing infections of any tissue, including lung tissue. Numerous bacterial, fungal,
parasitic, and viral infections also directly lead to hemorrhagic lesions or are present
together with bloody secretions. In addition, microbial TLR agonists are present with
hemoglobin in circulating blood in blood vessels if there is even minor hemolysis during
bacteremia, parasitemia, fungemia, or viremia. It is difficult to estimate the concentrations of
available free hemoglobin or heme in such infected tissues, drainage fluids, or blood. The
total concentration of hemoglobin in red blood cells in whole blood is ~150 mg/mL. We
found significant synergy with several TLR agonists (LPS, Pam3Cys, and Poly I:C) at 30
μg/mL, which is 1/5000th of this concentration. Estimation of the effective concentration of
heme is even more difficult. The approximate total heme concentration in red blood cells in
whole blood is approximately 9 mmol/L. We found significant synergy with 1 and 3 μmol/L,
which is approximately 1/3000th of this concentration. However, much of the free
hemoglobin and heme that is released from damaged red blood cells is likely to be bound
and blocked by haptoglobin [42,43] or by Hx [32] (Figure 6).
One of our most striking results was the suppressive effect of the Hx on both heme and
hemoglobin-LPS synergy. The suppression of the synergy of hemoglobin with LPS was
unexpected. There may be several potential mechanisms involved. First, Hx might simply
sequester free heme from interaction with macrophages. Indeed, this might also explain the
suppression of synergy with hemoglobin as well, because heme can be directly transferred
from hemoglobin to Hx [44]. Alternatively, heme-Hx complexes might induce
hemoxygenase-1, which has anti-inflammatory effects through its enzymatic products
carbon monoxide and biliverdin [45–47]. Finally, some other unknown mechanisms could
be involved. Elucidation of the mechanisms for the suppression of hemoglobin synergy with
LPS by Hx will need more studies.
A next step in evaluating the relevance of the findings will be to study the synergy of
hemoglobin and heme in some sort of in vivo system. Unfortunately, choosing an
appropriate model system that is reflective of human disease may be challenging [48].
Intrinsically, mice are multiple orders of magnitude more resilient to TLR4 agonist
challenge than humans, and the same relationship likely is true for other TLR agonists [23].
Therefore, assessment of the synergistic inflammation caused by blood products and
microbes might be better performed in a species that is similar to humans in sensitivity, such
as rabbits [23].
In summary, our studies suggest that hemoglobin and/or heme could contribute substantially
to the initiation of the inflammatory response induced by different microbial TLR agonists
in settings where trace amounts of blood coexist with bacterial or viral infection. The latter
is of special relevance given the current epidemic of H1N1 influenza and the knowledge that
severe disease can be accompanied by blood in the alveolar spaces [49]. Further work will
be needed to evaluate whether local concentrations of Hx are depleted in infected tissue
microenvironments and whether infusion of Hx to bind, neutralize, or clear hemoglobin or
hemin could be beneficial in some settings.
Acknowledgments
Financial support: National Institutes of Health (grants AI059010 and GM59694) and the Shriners Hospital for
Crippled Children (grant 8720).
Lin et al. Page 7
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
References
1. Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired
immunity. Nat Immunol. 2001; 2:675–80. [PubMed: 11477402]
2. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006; 124:783–
801. [PubMed: 16497588]
3. Bone RC, Balk RA, Cerra FB, et al. Definitions for sepsis and organ failure and guidelines for the
use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee.
American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992; 101:1644–
55. [PubMed: 1303622]
4. Rittirsch D, Flierl MA, Ward PA. Harmful molecular mechanisms in sepsis. Nat Rev Immunol.
2008; 8:776–87. [PubMed: 18802444]
5. Lombardi V, Van Overtvelt L, Horiot S, Moingeon P. Human dendritic cells stimulated via TLR7
and/or TLR8 induce the sequential production of Il-10, IFN-γ, and IL-17A by naive CD4+ T cells. J
Immunol. 2009; 182:3372–9. [PubMed: 19265114]
6. Strandskog G, Skjaeveland I, Ellingsen T, Jorgensen JB. Double-stranded RNA- and CpG DNA-
induced immune responses in Atlantic salmon: comparison and synergies. Vaccine. 2008; 26:4704–
15. [PubMed: 18602433]
7. Vanhoutte F, Paget C, Breuilh L, et al. Toll-like receptor (TLR)2 and TLR3 synergy and cross-
inhibition in murine myeloid dendritic cells. Immunol Lett. 2008; 116:86–94. [PubMed: 18166232]
8. He H, Genovese KJ, Nisbet DJ, Kogut MH. Synergy of CpG oligo-deoxynucleotide and double-
stranded RNA (poly I:C) on nitric oxide induction in chicken peripheral blood monocytes. Mol
Immunol. 2007; 44:3234–42. [PubMed: 17339052]
9. Roelofs MF, Joosten LA, Abdollahi-Roodsaz S, et al. The expression of toll-like receptors 3 and 7
in rheumatoid arthritis synovium is increased and costimulation of Toll-like receptors 3, 4, and 7/8
results in synergistic cytokine production by dendritic cells. Arthritis Rheum. 2005; 52:2313–22.
[PubMed: 16052591]
10. Napolitani G, Rinaldi A, Bertoni F, Sallusto F, Lanzavecchia A. Selected Toll-like receptor agonist
combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nat
Immunol. 2005; 6:769–76. [PubMed: 15995707]
11. Bagchi A, Herrup EA, Warren HS, et al. MyD88-dependent and MyD88-independent pathways in
synergy, priming, and tolerance between TLR agonists. J Immunol. 2007; 178:1164–71. [PubMed:
17202381]
12. Paul-Clark MJ, Sorrentino R, Bailey LK, Sriskandan S, Mitchell JA. Gram-positive and gram-
negative bacteria synergize with oxidants to release CXCL8 from innate immune cells. Mol Med.
2008; 14:238–46. [PubMed: 18231574]
13. Tsujimoto H, Ono S, Efron PA, Scumpia PO, Moldawer LL, Mochizuki H. Role of Toll-like
receptors in the development of sepsis. Shock. 2008; 29:315–21. [PubMed: 18277854]
14. Bornside GH, Bouis PJ Jr, Cohn I Jr. Hemoglobin and Escherichia coli, a lethal intraperitoneal
combination. J Bacteriol. 1968; 95:1567–71. [PubMed: 4870275]
15. Roth RI, Levin FC, Levin J. Distribution of bacterial endotoxin in human and rabbit blood and
effects of stroma-free hemoglobin. Infect Immun. 1993; 61:3209–15. [PubMed: 8335351]
16. Su D, Roth RI, Yoshida M, Levin J. Hemoglobin increases mortality from bacterial endotoxin.
Infect Immun. 1997; 65:1258–66. [PubMed: 9119460]
17. Su D, Roth RI, Levin J. Hemoglobin infusion augments the tumor necrosis factor response to
bacterial endotoxin (lipopolysaccharide) in mice. Crit Care Med. 1999; 27:771–8. [PubMed:
10321668]
18. Bodet C, Chandad F, Grenier D. Hemoglobin and LPS act in synergy to amplify the inflammatory
response. J Dent Res. 2007; 86:878–82. [PubMed: 17720859]
19. Kaca W, Roth RI, Levin J. Hemoglobin, a newly recognized lipopolysaccharide (LPS)-binding
protein that enhances LPS biological activity. J Biol Chem. 1994; 269:25078–84. [PubMed:
7929195]
Lin et al. Page 8
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
20. Brandenburg K, Garidel P, Andra J, et al. Cross-linked hemoglobin converts endotoxically inactive
pentaacyl endotoxins into a physiologically active conformation. J Biol Chem. 2003; 278:47660–
9. [PubMed: 13679376]
21. Bornside GH, Bouis PJ Jr, Cohn I Jr. Enhancement of Escherichia coli infection and endotoxic
activity by hemoglobin and ferric ammonium citrate. Surgery. 1970; 68:350–5. [PubMed:
4915899]
22. Su D, Roth RI, Levin J. Hemoglobin infusion augments the tumor necrosis factor response to
bacterial endotoxin (lipopolysaccharide) in mice. Crit Care Med. 1999; 27:771–8. [PubMed:
10321668]
23. Warren HS, Fitting C, Hoff E, et al. Resilience to bacterial infection: difference between species
could be due to proteins in serum. J Infect Dis. 2010; 201:223–32. [PubMed: 20001600]
24. Figueiredo RT, Fernandez PL, Mourao-Sa DS, et al. Characterization of heme as activator of Toll-
like receptor 4. J Biol Chem. 2007; 282:20221–9. [PubMed: 17502383]
25. Kawai T, Adachi O, Ogawa T, Takeda K, Akira S. Unresponsiveness of MyD88-deficient mice to
endotoxin. Immunity. 1999; 11:115–22. [PubMed: 10435584]
26. Andrade CT, Barros LA, Lima MC, Azero EG. Purification and characterization of human
hemoglobin: effect of the hemolysis conditions. Int J Biol Macromol. 2004; 34:233–40. [PubMed:
15374679]
27. Liang X, Lin T, Sun G, Beasley-Topliffe L, Cavaillon JM, Warren HS. Hemopexin down-regulates
LPS-induced proinflammatory cytokines from macrophages. J Leukoc Biol. 2009; 86:229–35.
[PubMed: 19395472]
28. Novitsky TJ, Roslansky PF, Siber GR, Warren HS. Turbidometric method for quantifying serum
inhibition of limulus amoebocyte lysate response. J Clin Micro. 1985; 20:211–6.
29. Schilling D, Thomas K, Nixdorff K, Vogel SN, Fenton MJ. Toll-like receptor 4 and Toll–IL-1
receptor domain-containing adapter protein (TIRAP)/myeloid differentiation protein 88 adapter-
like (Mal) contribute to maximal IL-6 expression in macrophages. J Immunol. 2002; 169:5874–80.
[PubMed: 12421970]
30. Liang MD, Bagchi A, Warren HS, et al. Bacterial peptidoglycan-associated lipoprotein: a naturally
occurring Toll-like receptor 2 agonist that is shed into serum and has synergy with
lipopolysaccharide. J Infect Dis. 2005; 191:939–48. [PubMed: 15717270]
31. Yang H, Wang H, Bernik TR, et al. Globin attenuates the innate immune response to endotoxin.
Shock. 2002; 17:485–90. [PubMed: 12069185]
32. Tolosano E, Altruda F. Hemopexin: structure, function, and regulation. DNA Cell Biol. 2002;
21:297–306. [PubMed: 12042069]
33. Paoli M, Anderson BF, Baker HM, Morgan WT, Smith A, Baker EN. Crystal structure of
hemopexin reveals a novel high-affinity heme site formed between two beta-propeller domains.
Nat Struct Biol. 1999; 6:926–31. [PubMed: 10504726]
34. Watanabe J, Grijalva V, Hama S, et al. Hemoglobin and its scavenger protein haptoglobin
associate with ApoA-1–containing particles and influence the inflammatory properties and
function of high density lipoprotein. J Biol Chem. 2009; 284:18292–301. [PubMed: 19433579]
35. Wilson BMG, Severn A, Rapson NT, Chana J, Hopkins P. A convenient human whole blood
culture system for studying the regulation of tumour necrosis factor release by bacterial
lipopolysaccharide. J Immunol Methods. 1991; 139:233–40. [PubMed: 1904465]
36. Cox KH, Ofek I, Hasty DL. Enhancement of macrophage stimulation by lipoteichoic acid and the
costimulant hemoglobin is dependent on Toll-like receptors 2 and 4. Infect Immun. 2007;
75:2638–41. [PubMed: 17296755]
37. Tanabe S, Gottschalk M, Grenier D. Hemoglobin and Streptococcus suis cell wall act in synergy to
potentiate the inflammatory response of monocyte-derived macrophages. Innate Immun. 2008;
14:357–63. [PubMed: 19039059]
38. Takeda, K.; Akira, S. Toll-like receptors. In: Coligan, JE., et al., editors. Current Protocols in
Immunology. Vol. chapter 14. 2007.
39. Takeda K, Akira S. Toll-like receptors in innate immunity. Int Immunol. 2005; 17:1–14. [PubMed:
15585605]
Lin et al. Page 9
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
40. Crowley JP, Metzger J, Gray A, Pivacek LE, Cassidy G, Valeri CR. Infusion of stroma-free cross-
linked hemoglobin during acute gram-negative bacteremia. Circ Shock. 1993; 41:144–9.
[PubMed: 8269643]
41. Bogdan C. Nitric oxide and the immune response. Nat Immunol. 2001; 2:907–16. [PubMed:
11577346]
42. Schaer DJ, Alayash AI, Buehler PW. Gating the radical hemoglobin to macrophages: the anti-
inflammatory role of CD163, a scavenger receptor. Antioxid Redox Signal. 2007; 9:991–9.
[PubMed: 17508920]
43. Polticelli F, Bocedi A, Minervini G, Ascenzi P. Human haptoglobin structure and function—a
molecular modelling study. FEBS J. 2008; 275:5648–56. [PubMed: 18959750]
44. Hrkal Z, Vodrazka Z, Kalousek I. Transfer of heme from ferrihemoglobin and ferrihemoglobin
isolated chains to hemopexin. Eur J Biochem. 1974; 43:73–8. [PubMed: 4209590]
45. Chung SW, Liu X, Macias AA, Baron RM, Perrella MA. Heme oxygenase-1-derived carbon
monoxide enhances the host defense response to microbial sepsis in mice. J Clin Invest. 2008;
118:239–47. [PubMed: 18060048]
46. Otterbein LE, Soares MP, Yamashita K, Bach FH. Heme oxygenase-1: unleashing the protective
properties of heme. Trends Immunol. 2003; 24:449–55. [PubMed: 12909459]
47. Pamplona A, Ferreira A, Balla J, et al. Heme oxygenase-1 and carbon monoxide suppress the
pathogenesis of experimental cerebral malaria. Nat Med. 2007; 13:703–10. [PubMed: 17496899]
48. Warren HS. Editorial: Mouse models to study sepsis syndrome in humans. J Leukoc Biol. 2009;
86:199–201. [PubMed: 19643738]
49. Perez-Padilla R, Rosa-Zamboni D, Ponce DL, et al. Pneumonia and respiratory failure from swine-
origin influenza A (H1N1) in Mexico. N Engl J Med. 2009; 361:680–9. [PubMed: 19564631]
Lin et al. Page 10
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1.
Synergy of hemoglobin (Hb) with lipopolysaccharide (LPS), Pam3Cys, Poly I:C, loxoribine,
and bacterial DNA (CpG). Bone marrow–derived macrophages from C57BL/6 mice were
washed 3 times with serum-free medium and then were cultured overnight with either Hb
alone or Hb with various Toll-like receptor agonists: LPS (1 ng/mL) (A and B ), Pam3Cys
(P3C; 10 ng/mL) (C and D ), Poly I:C (150 μg/mL) (E and F ), loxoribine (250 μmol/L) (G
and H ), and CpG (0.1 μmol/L) (I and J ). Concentrations of tumor necrosis factor (TNF) (A,
C, E, G, and I ) and interleukin (IL)–6 (B, D, F, H, and J ) in the supernatants were
determined by enzyme-linked immunosorbent assay. The results denote the mean ± standard
error and are representative of 4 independent experiments. *P <.05 and **P <.01, compared
between cells treated with and without Hb.
Lin et al. Page 11
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 2.
Synergy of hemoglobin (Hb) with killed Escherichia coli and Staphylococcus aureus. Bone
marrow–derived macrophages from C57BL/6 mice were washed 3 times with serum-free
medium and then cultured overnight (A, B, E, and F ) or at different times (C, D, G, and H )
with heat-killed E. coli at indicated concentrations (A and B ) or at 105 cfu/mL (C and D ) or
with S. aureus at indicated concentrations (E and F ) or at 107 cfu/mL (G and H ) in the
absence or presence of Hb at 300 μg/mL. Concentrations of tumor necrosis factor (TNF) (A,
C, E, and G ) and interleukin (IL)–6 (B, D, F, and H ) in the supernatants were determined
by enzyme-linked immunosorbent assay. The results denote the mean ± standard error and
Lin et al. Page 12
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
are representative of 4 independent experiments. *P <.05, **P <.01, ***P <.001, compared
between cells treated with and without Hb.
Lin et al. Page 13
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 3.
Synergy of hemoglobin (Hb) with Toll-like receptor (TLR) agonists is not TLR4 dependent.
Bone marrow–derived macrophages from HeN and HeJ mice were washed 3 times with
serum-free medium and then cultured overnight with TLR agonists alone, including
Pam3Cys (P3C; 10 ng/mL) (A and B ), Poly I:C (P[I:C]; 150 μg/mL) (C and D ), loxoribine
(Lox; 250 μmol/L) (E and F ), and bacterial DNA (CpG; 0.1 μmol/L) (G and H ), or with Hb
at 100 μg/mL (A–D ) or 300 μg/mL (E and F ) or 1000 μg/mL (G and H ) indicated
concentrations. Concentrations of tumor necrosis factor (TNF) (A, C, E, and G ) and
interleukin (IL)–6 (B, D, F, and H ) in the supernatants were determined by enzyme-linked
immunosorbent assay. Each graph denotes the percentage of cytokine production when
cytokine production by TLR agonist alone is assigned to 100%. Results denote the mean ±
the standard error and are representative of 4 independent experiments. *P <.05 and **P <.
01, compared between cells treated with and without Hb.
Lin et al. Page 14
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 4.
Synergy of hemoglobin (Hb) with lipopolysaccharide (LPS) and Poly I:C is not MyD88
dependent. Bone marrow–derived macrophages from C57BL/6 and MyD88 knockout
(MyD88KO) mice were washed 3 times with serum-free medium and then cultured with
Toll-like receptor (TLR) agonists LPS (1 ng/mL) (A) or Poly I:C (P[I:C]; 150 μg/mL) (B )
and Hb at 100 μg/mL. Concentrations of tumor necrosis factor (TNF) in the supernatants
were determined by enzyme-linked immunosorbent assay. The data denote the percentage of
cytokine production, when cytokine production by TLR agonist alone is assigned to 100%.
The results denote the mean ± the standard error and are representative of 3 independent
experiments. *P <.05 and **P <.01, compared between cells treated with and without Hb.
Lin et al. Page 15
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 5.
Synergy of hemin with lipopolysaccharide (LPS), Poly I:C, and loxoribine, and the role of
Toll-like receptor (TLR)–4 and MyD88. Bone marrow–derived macrophages from C57BL/
6, C3H/HeN, C3H/HeJ, or MyD88 knockout (MyD88KO) mice were washed 3 times with
serum-free medium and then cultured with TLR agonists. A, C57BL/6 cells, hemin
concentrations as shown (the open bar denotes no hemin; vertical lines, 1 μmol/L hemin; and
the solid bar, 3 μmol/L hemin) and, where indicated, LPS (1 ng/mL). The tumor necrosis
factor (TNF) concentrations in the culture are shown. B and C, C3H/HeN (HeN) and C3H/
HeJ (HeJ) cells cultured with Poly I:C (P[I:C], 150 μg/mL) (B ) or loxoribine (Lox; 250
μmol/L) (C ) in the absence or presence of hemin (3 μmol/L). D and E, C57BL/6 or MyD88
KO cells cultured with Poly I:C (P[I:C]; 150 μg/mL) (D ) or LPS (1 ng/mL) in the absence
or presence of hemin (3 μmol/L) (E ). B–E, The percentage of TNF production when TNF
by the same type of cells stimulated by TLR agonist alone is assigned to 100%. The results
denote the mean ± the standard error and are representative of four independent experiments.
*P <.05, **P <.01, and ***P <.001, compared between cells treated with and without
hemin.
Lin et al. Page 16
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 6.
Effect of mouse or human hemopexin (mHx or hHx) on hemin or hemoglobin (Hb) synergy
with lipopolysaccharide (LPS) or Escherichia coli on bone marrow–derived macrophages
(BMDMs) or human whole blood. BMDMs from C57BL/6 mice were washed 3 times with
serum-free medium and then cultured overnight with 1 ng/mL of LPS in the absence () or
presence () of 300 μg/mL mHx and indicated concentrations of hemin (A) or Hb (B ).
Concentrations of tumor necrosis factor (TNF) in the supernatants were determined by
enzyme-linked immunosorbent assay. Fresh human whole blood was diluted 1:4 in serum-
free medium and cultured for 1 h and then were coincubated overnight with 2.5 ng/mL LPS
or 105 cfu/mL E. coli in the absence or presence of mHx (300 μg/mL) (C ), or 105 cfu/mL of
E. coli with or without hHx (400 μg/mL) (D ). The results denote the mean ± the standard
error and are representative of more than 4 independent experiments. *P <.05, **P <.01,
***P <.001, compared between samples treated with and without Hx.
Lin et al. Page 17
J Infect Dis. Author manuscript; available in PMC 2011 August 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... In 2007, heme was demonstrated to mediate proinflammatory signalling in mouse macrophages via the CD14-TLR4-MyD88 axis, which suggested heme as a TLR4 agonist [37]. Up to date, the involvement of several cofactors (CD14, MD2), adaptor, and effector proteins in heme-triggered TLR4 signalling in macrophages, platelets, HEK293, peripheral blood mononuclear (PBMCs), microglial, dendritic, and endothelial cells has been described [14,16,17,20,[37][38][39][40][41][42][43][44], which was recently contextualized in the knowledge graph 'HemeKG' [19]. Corroborating the heme-TLR4 association, stimulating cells with heme induces signalling via NF-κB and AP1 resulting in the expression of proinflammatory mediators, such as TNF and the functional IL-8 homologue CXCL1 [37,39,45]. ...
... Up to date, the involvement of several cofactors (CD14, MD2), adaptor, and effector proteins in heme-triggered TLR4 signalling in macrophages, platelets, HEK293, peripheral blood mononuclear (PBMCs), microglial, dendritic, and endothelial cells has been described [14,16,17,20,[37][38][39][40][41][42][43][44], which was recently contextualized in the knowledge graph 'HemeKG' [19]. Corroborating the heme-TLR4 association, stimulating cells with heme induces signalling via NF-κB and AP1 resulting in the expression of proinflammatory mediators, such as TNF and the functional IL-8 homologue CXCL1 [37,39,45]. However, despite triggering a signalling response dependent on TLR4, heme is unable to induce the expression of TRIF-dependent genes on macrophages [37,45]. ...
... However, despite triggering a signalling response dependent on TLR4, heme is unable to induce the expression of TRIF-dependent genes on macrophages [37,45]. Moreover, heme synergizes with several PAMPs in a mechanism independent of TLR4, MyD88 and TRIF, but dependent of SYK and ROS [39,46]. ...
Article
Full-text available
Haemolytic disorders, such as sickle cell disease, are accompanied by the release of high amounts of labile heme into the intravascular compartment resulting in the induction of proinflammatory and prothrombotic complications in affected patients. In addition to the relevance of heme-regulated proteins from the complement and blood coagulation systems, activation of the TLR4 signalling pathway by heme was ascribed a crucial role in the progression of these pathological processes. Heme binding to the TLR4-MD2 complex has been proposed recently, however, essential mechanistic information of the processes at the molecular level, such as heme-binding kinetics, the heme-binding capacity and the respective heme-binding sites (HBMs) is still missing. We report the interaction of TLR4, MD2 and the TLR4-MD2 complex with heme and the consequences thereof by employing biochemical, spectroscopic, bioinformatic and physiologically relevant approaches. Heme binding occurs transiently through interaction with up to four HBMs in TLR4, two HBMs in MD2 and at least four HBMs in their complex. Functional studies highlight that mutations of individual HBMs in TLR4 preserve full receptor activation by heme, suggesting that heme interacts with TLR4 through different binding sites independently of MD2. Furthermore, we confirm and extend the major role of TLR4 for heme-mediated cytokine responses in human immune cells.
... Acting as the prosthetic group for such hemoproteins as haemoglobin, myoglobin and cytochrome P450, heme plays an essential role in a range of biological processes, which include oxygen transport and storage, electron transfer and cellular metabolism (94). However, when released from hemoproteins, unbound heme promotes oxidative stress, inflammation and tissue injury via its ability to catalyse the formation of highly reactive oxygen free radicals, amplify TLRmediated immune responses and activate the complement cascade (94)(95)(96)(97)(98). Thus, in the setting of critical illness, links have been suggested between elevated concentrations of cell-free heme, the SIRS response and organ injury (99). ...
Article
Full-text available
Associated with the development of hospital-acquired infections, major traumatic injury results in an immediate and persistent state of systemic immunosuppression, yet the underlying mechanisms are poorly understood. Detected in the circulation in the minutes, days and weeks following injury, damage associated molecular patterns (DAMPs) are a heterogeneous collection of proteins, lipids and DNA renowned for initiating the systemic inflammatory response syndrome. Suggesting additional immunomodulatory roles in the post-trauma immune response, data are emerging implicating DAMPs as potential mediators of post-trauma immune suppression. Discussing the results of in vitro, in vivo and ex vivo studies, the purpose of this review is to summarise the emerging immune tolerising properties of cytosolic, nuclear and mitochondrial-derived DAMPs. Direct inhibition of neutrophil antimicrobial activities, the induction of endotoxin tolerance in monocytes and macrophages, and the recruitment, activation and expansion of myeloid derived suppressor cells and regulatory T cells are examples of some of the immune suppressive properties assigned to DAMPs so far. Crucially, with studies identifying the molecular mechanisms by which DAMPs promote immune suppression, therapeutic strategies that prevent and/or reverse DAMP-induced immunosuppression have been proposed. Approaches currently under consideration include the use of synthetic polymers, or the delivery of plasma proteins, to scavenge circulating DAMPs, or to treat critically-injured patients with antagonists of DAMP receptors. However, as DAMPs share signalling pathways with pathogen associated molecular patterns, and pro-inflammatory responses are essential for tissue regeneration, these approaches need to be carefully considered in order to ensure that modulating DAMP levels and/or their interaction with immune cells does not negatively impact upon anti-microbial defence and the physiological responses of tissue repair and wound healing.
... Vinci et al. observed in sickle cell mice that the administration of human exogenous hemopexin attenuates the inflammatory phenotype of macrophages [18]. Thus, our findings are in line with recent studies demonstrating the role of heme in macrophage polarization towards a pro-inflammatory phenotype leading to the activation of inflammatory cytokines via TLR-4 stimulation [20][21][22]71]. ...
Article
Full-text available
Chronic widespread pain (CWP) is associated with a high rate of disability and decreased quality of life in people with HIV-1 (PWH). We previously showed that PWH with CWP have increased hemolysis and elevated plasma levels of cell-free heme, which correlate with low endogenous opioid levels in leukocytes. Further, we demonstrated that cell-free heme impairs β-endorphin synthesis/release from leukocytes. However, the cellular mechanisms by which heme dampens β-endorphin production are inconclusive. The current hypothesis is that heme-dependent TLR4 activation and macrophage polarization to the M1 phenotype mediate this phenomenon. Our novel findings showed that PWH with CWP have elevated M1-specific macrophage chemokines (ENA-78, GRO-α, and IP-10) in plasma. In vitro, hemin-induced polarization of M0 and M2 macrophages to the M1 phenotype with low β-endorphins was mitigated by treating cells with the TLR4 inhibitor, TAK-242. Similarly, in vivo phenylhydrazine hydrochloride (PHZ), an inducer of hemolysis, injected into C57Bl/6 mice increased the M1/M2 cell ratio and reduced β-endorphin levels. However, treating these animals with the heme-scavenging protein hemopexin (Hx) or TAK-242 reduced the M1/M2 ratio and increased β-endorphins. Furthermore, Hx attenuated heme-induced mechanical, heat, and cold hypersensitivity, while TAK-242 abrogated hypersensitivity to mechanical and heat stimuli. Overall, these results suggest that heme-mediated TLR4 activation and M1 polarization of macrophages correlate with impaired endogenous opioid homeostasis and hypersensitivity in people with HIV.
... In addition, reactive oxygen species are generated through Haber-Weiss-and Fenton-reactions mediated by hemoglobin, heme and iron [15,16]. CFH can also cause direct cytotoxic injury to cell membranes, plasma proteins and lipids [11,[17][18][19]. Furthermore, elevated plasma concentrations of CFH have been observed to be associated with direct organ injuries, such as renal failure, intestinal mucosal damage, or lung injury [2,4,20,21]. ...
Article
Full-text available
Background: Hemolysis is associated with increased mortality in patients with sepsis, ARDS, or therapy with extracorporeal membrane oxygenation (ECMO). To quantify a critical threshold of hemolysis in patients with ARDS and treatment with veno-venous ECMO, we aimed to identify cutoff values for cell-free hemoglobin (CFH) and haptoglobin (Hp) plasma concentrations associated with a significant increase in ICU mortality. Methods: Patients with ARDS admitted to a tertiary ARDS referral center between 01/2007 and 12/2018 and treatment with veno-venous ECMO were included. Cutoff values for mean CFH (mCFH) and mean Hp (mHp) plasma concentrations dividing the cohort into groups with significantly different ICU mortalities were calculated and patient characteristics were compared. A multiple logistic regression model with stepwise backward variable selection was included. In addition, cutoff values for vulnerable relative timespans for the respective CFH and Hp concentrations were calculated. Results: A quantitative cutoff value of 11 mg/dl for mCFH separated the cohort (n = 442) regarding ICU mortality (mCFH ≤ 11 mg/dl: 38%, [95%-CI: 32.22-43.93] (n = 277) vs. mCFH > 11 mg/dl: 70%, [61.99-76.47] (n = 165), p < 0.001). Analogously, a mHp cutoff value ≤ 0.39 g/l was associated with a significant increase in ICU mortality (mHp ≤ 0.39 g/l: 68.7%, [60.91-75.61] (n = 163) vs. mHp > 0.39 g/l: 38.7%, [33.01-44.72] (n = 279), p < 0.001). The independent association of ICU mortality with CFH and Hp cutoff values was confirmed by logistic regression adjusting for confounders (CFH Grouping: OR 3.77, [2.51-5.72], p < 0.001; Hp Grouping: OR 0.29, [0.19-0.43], p < 0.001). A significant increase in ICU mortality was observed when CFH plasma concentration exceeded the limit of 11 mg/dl on 13.3% of therapy days (≤ 13.3% of days with CFH > 11 mg/dl: 33%; [26.81-40.54] (n = 192) vs. > 13.3% of days with CFH > 11 mg/dl: 62%; [56.05-68.36] (n = 250), p < 0.001). Analogously, a mortality increase was detected when Hp plasma concentration remained ≤ 0.39 g/l for > 18.2% of therapy days (≤ 18.2% days with Hp ≤ 0.39 g/l: 27%; [19.80-35.14] (n = 138) vs. > 18.2% days with Hp ≤ 0.39 g/l: 60%; [54.43-65.70] (n = 304), p < 0.001). Conclusions: Moderate hemolysis with mCFH-levels as low as 11 mg/dl impacts mortality in patients with ARDS and therapy with veno-venous ECMO. Furthermore, a cumulative dose effect should be considered indicated by the relative therapy days with CFH-concentrations > 11 mg/dl. In addition, also Hp plasma concentrations need consideration when the injurious effect of elevated CFH is evaluated.
... It seems logical to hypothesize that, if this protein is sequestered in whole or in part by HNE, there could be less of it available to defend the organism from the attack of the virus. Hemopexin (HPX) is a protein whose synthesis is induced after an inflammatory event [56] and that has a high affinity for the heme group that, among other features, amplifies the innate immune responses induced by toll-like receptor 4 causing adverse immune reactions such as the migration of leukocytes and the production of adhesion molecules and cytokines [57]. In the pulmonary system, this results in increased vascular permeability, interstitial edema, and migration of inflammatory cells [58]. ...
Article
Full-text available
Neutrophils play a pathogenic role in COVID-19 by releasing Neutrophils Extracellular Traps (NETs) or human neutrophil elastase (HNE). Given that HNE is inhibited by α1-antitrypsin (AAT), we aimed to assess the content of HNE, α1-antitrypsin (AAT) and HNE–AAT complexes (the AAT/HNE balance) in 33 bronchoalveolar lavage fluid (BALf) samples from COVID-19 patients. These samples were submitted for Gel-Electrophoresis, Western Blot and ELISA, and proteins (bound to AAT or HNE) were identified by Liquid Chromatography-Mass Spectrometry. NETs’ release was analyzed by confocal microscopy. Both HNE and AAT were clearly detectable in BALf at high levels. Contrary to what was previously observed in other settings, the formation of HNE–AAT complex was not detected in COVID-19. Rather, HNE was found to be bound to acute phase proteins, histones and C3. Due to the relevant role of NETs, we assessed the ability of free AAT to bind to histones. While confirming this binding, AAT was not able to inhibit NET formation. In conclusion, despite the finding of a high burden of free and bound HNE, the lack of the HNE–AAT inhibitory complex in COVID-19 BALf demonstrates that AAT is not able to block HNE activity. Furthermore, while binding to histones, AAT does not prevent NET formation nor their noxious activity.
... For example, viral infection that coincides with intravascular hemolysis or damage to RBCs results in the release of intracellular stores of hemoglobin and heme into the bloodstream or promote erythrophagocytosis by tissue resident macrophages (especially in the spleen and liver), which in turn can augment the inflammatory response and promote tissue damage, sepsis, and thrombosis. [28][29][30] Infection could also lead to an increased production of cytokines by cells in these local environments, thereby facilitating additional interactions with RBCs to support pathological alterations such as impaired maternofetal gas exchange, 31 hypercoagulation due to RBC agglutination and eryptosis 32,33 and ROSmediated damage to RBC membrane components. 34,35 Functional consequences of altering the membrane lipid composition of RBCs include an accumulation of lipid oxidation products, 36 compromised oxygen and ion transport 37 and changes to the placental transfer of fatty acids. ...
Article
Full-text available
Background: Diseases caused by arthropod-borne viruses remain a burden to global health; in particular, Zika virus (ZIKV) has been reported in 87 countries and territories. In healthy blood donors, ZIKV RNA can be detected in red blood cells (RBCs) months after infection, clearance of detectable nucleic acid in plasma, and seroconversion. However, little information is available on the impact of ZIKV infection to metabolism. Study design and methods: We applied mass spectrometry-based metabolomics and lipidomics approaches to investigate the impact of ZIKV infection on RBCs over the course of infection. ZIKV-infected blood donors (n = 25) were identified through molecular and serologic methods, which included nucleic acid amplification testing and real-time polymerase chain reaction (PCR) for detection of ZIKV RNA and enzyme-linked immunosorbent assay (ELISA) for detection of flavivirus-specific IgM and IgG. Results: In ZIKV RNA-positive donors, we observed lower glucose and lactate levels, and higher levels of ribose phosphate, suggestive of the activation of the pentose phosphate pathway. The top pathways altered in RBCs from ZIKV-IgM-positive donors include amino acid metabolism and biosynthesis, fatty acid metabolism and biosynthesis, linoleic acid and arachidonate metabolism and glutathione metabolism. RBCs from ZIKV-infected donors had increased levels of early glycolytic metabolites, and higher levels of metabolites of the pentose phosphate pathway. Alterations in acyl-carnitine and fatty acid metabolism are consistent with impaired membrane lipid homeostasis in RBCs from ZIKV IgM positive donors. Conclusion: RBC from healthy blood donors who had been infected by ZIKV are characterized by long-lasting metabolic alterations even months after infection has resolved.
... The mechanisms of toxicity caused by CFH include nitric oxide scavenging with concomitant vasoconstriction, platelet aggregation, inflammation, lipid peroxidation, mitochondrial damage, increased oxidative stress, and stimulation of pro-inflammatory receptors [7][8][9][10]. While renal damage is commonly attributed to general or regional renal hypoperfusion, increased levels of circulating CFH lead to glomerular filtration of CFH which may further aggravate renal damage by subsequent tubular injury [6]. ...
Article
Full-text available
Abstract Background Increased plasma concentrations of circulating cell-free hemoglobin (CFH) are supposed to contribute to the multifactorial etiology of acute kidney injury (AKI) in critically ill patients while the CFH-scavenger haptoglobin might play a protective role. We evaluated the association of CFH and haptoglobin with AKI in patients with an acute respiratory distress syndrome (ARDS) requiring therapy with VV ECMO. Methods Patients with CFH and haptoglobin measurements before initiation of ECMO therapy were identified from a cohort of 1044 ARDS patients and grouped into three CFH concentration groups using a risk stratification. The primary objective was to assess the association of CFH and haptoglobin with KDIGO stage 3 AKI. Further objectives included the identification of a target haptoglobin concentration to protect from CFH-associated AKI. Measurements and main results Two hundred seventy-three patients fulfilled the inclusion criteria. Of those, 154 patients (56.4%) had AKI at ECMO initiation. The incidence of AKI increased stepwise with increasing concentrations of CFH reaching a plateau at 15 mg/dl. Compared to patients with low [
Article
The strength and duration of the NF-κB signaling response must be tightly modulated to avoid an inadequate or excessive immune response. Relish, a core NF-κB transcription factor of the Drosophila Imd pathway, can control the expression of antimicrobial peptides, including Dpt and AttA, to defend against Gram-negative bacterial infections, but whether Relish may regulate miRNA expression to participate in the immune response remains unclear. In this study, taking advantage of Drosophila S2 cells and different overexpression/knockout/knockdown flies, we first found that Relish could directly activate the expression of miR-308 to negatively regulate the immune response and promote the survival of Drosophila during Enterobacter cloacae infection. Second, our results demonstrated that Relish-mediated expression of miR-308 could suppress target gene Tab2 to attenuate the Drosophila Imd pathway signal during the middle and late stages of the immune response. Third, we detected the dynamic expression patterns of Dpt, AttA, Relish, miR-308, and Tab2 in wild-type flies after E. coli infection, which further revealed that the feedback regulatory loop of Relish-miR-308-Tab2 plays a crucial role in the immune response and homeostasis maintenance of the Drosophila Imd pathway. Overall, our present study not only illustrates an important mechanism by which this Relish-miR-308-Tab2 regulatory axis can negatively control the Drosophila immune response and participate in homeostasis maintenance but also provides new insights into the dynamic regulation of the NF-κB/miRNA expression network of animal innate immunity.
Article
Full-text available
Soil-transmitted helminth [mainly Strongyloidiasis stercoralis (Ss)] and tuberculous lymphadenitis (TBL) coinfection in humans is a significant public health problem. We have previously shown that TBL+Ss+ coinfection significantly alters diverse cytokine, matrix metalloproteinase, and tissue inhibitors of metalloproteinase profiles. However, no data is available to understand the influence of Ss coinfection in TBL disease with respect to iron status biomarkers. Hence, we have studied the effect of Ss coinfection on the circulating levels of iron status (ferritin, transferrin [TF], apotransferrin [ApoT], hepcidin, hemopexin) biomarkers in TBL disease. Our results show that TBL+Ss+ and/or TBL+Ss- individuals are associated with significantly altered biochemical and hematological (red blood cell (RBC) counts, hemoglobin (Hb), hematocrit (HCT), mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH) were decreased, and platelets were increased) parameters compared to TBL-Ss+ individuals. Our results also show that TBL+Ss+ coinfection is associated with diminished circulating levels of ferritin, ApoT, hepcidin, and hemopexin compared to TBL+Ss- individuals. TBL+Ss+ and TBL+Ss- groups are associated with altered iron status biomarkers (decreased ferritin [TBL+Ss+ alone] and increased TF, ApoT, hepcidin and hemopexin [TBL+Ss- alone]) compared to TBL-Ss+ group. The heat map expression profile and principal component analysis (PCA) analysis of iron status biomarkers were significantly altered in TBL+Ss+ compared to TBL+Ss- and/or TBL-Ss+ individuals. A significant correlation (positive/negative) was obtained among the biochemical and hematological parameters (white blood cells (WBC)/ferritin, TF, and hepcidin, mean corpuscular hemoglobin concentration (MCHC)/ferritin and hemopexin) with iron status biomarkers. Finally, receiver operating characteristic (ROC) analysis revealed that hemopexin was significantly associated with greater specificity and sensitivity in discriminating TBL+Ss+ and TBL+Ss- coinfected individuals. Thus, our data conclude that Ss coinfection is associated with altered iron status biomarkers indicating that coinfection might alter the host- Mtb interface and could influence the disease pathogenesis.
Article
The artificial lung (AL) technology is one of the membrane-based artificial organs that partly augments lung functions, i.e. blood oxygenation and CO2 removal. It is generally employed as an extracorporeal membrane oxygenation (ECMO) device to treat acute and chronic lung-failure patients, and the recent outbreak of the COVID-19 pandemic has re-emphasized the importance of this technology. The principal component in AL is the polymeric membrane oxygenator that facilitates the O2/CO2 exchange with the blood. Despite the considerable improvement in anti-thrombogenic biomaterials in other applications (e.g., stents), AL research has not advanced at the same rate. This is partly because AL research requires interdisciplinary knowledge in biomaterials and membrane technology. Some of the promising biomaterials with reasonable hemocompatibility — such as emerging fluoropolymers of extremely low surface energy — must first be fabricated into membranes to exhibit effective gas exchange performance. As AL membranes must also demonstrate high hemocompatibility in tandem, it is essential to test the membranes using in-vitro hemocompatibility experiments before in-vivo test. Hence, it is vital to have a reliable in-vitro experimental protocol that can be reasonably correlated with the in-vivo results. However, current in-vitro AL studies are unsystematic to allow a consistent comparison with in-vivo results. More specifically, current literature on AL biomaterial in-vitro hemocompatibility data are not quantitatively comparable due to the use of unstandardized and unreliable protocols. Such a wide gap has been the main bottleneck in the improvement of AL research, preventing promising biomaterials from reaching clinical trials. This review summarizes the current state-of-the-art and status of AL technology from membrane researcher perspectives. Particularly, most of the reported in-vitro experiments to assess AL membrane hemocompatibility are compiled and critically compared to suggest the most reliable method suitable for AL biomaterial research. Also, a brief review of current approaches to improve AL hemocompatibility is summarized. Statement of Significance The importance of Artificial Lung (AL) technology has been re-emphasized in the time of the COVID-19 pandemic. The utmost bottleneck in the current AL technology is the poor hemocompatibility of the polymer membrane used for O2/CO2 gas exchange, limiting its use in the long-term. Unfortunately, most of the in-vitro AL experiments are unsystematic, irreproducible, and unreliable. There are no standardized in-vitro hemocompatibility characterization protocols for quantitative comparison between AL biomaterials. In this review, we tackled this bottleneck by compiling the scattered in-vitro data and suggesting the most suitable experimental protocol to obtain reliable and comparable hemocompatibility results. To the best of our knowledge, this is the first review paper focusing on the hemocompatibility challenge of AL technology.
Article
Full-text available
The ubiquitous use of heme in animals poses severe biological and chemical challenges. Free heme is toxic to cells and is a potential source of iron for pathogens. For protection, especially in conditions of trauma, inflammation and hemolysis, and to maintain iron homeostasis, a high-affinity binding protein, hemopexin, is required. Hemopexin binds heme with the highest affinity of any known protein, but releases it into cells via specific receptors. The crystal structure of the heme–hemopexin complex reveals a novel heme binding site, formed between two similar four-bladed β-propeller domains and bounded by the interdomain linker. The ligand is bound to two histidine residues in a pocket dominated by aromatic and basic groups. Further stabilization is achieved by the association of the two β-propeller domains, which form an extensive polar interface that includes a cushion of ordered water molecules. We propose mechanisms by which these structural features provide the dual function of heme binding and release.
Article
Full-text available
Vertebrates vary in resistance and resilience to infectious diseases, and the mechanisms that regulate the tradeoff between these often opposing protective processes are not well understood. Variability in the sensitivity of species to the induction of damaging inflammation in response to equivalent pathogen loads (resilience) complicates the use of animal models that reflect human disease. We found that induction of proinflammatory cytokines from macrophages in response to inflammatory stimuli in vitro is regulated by proteins in the sera of species in inverse proportion to their in vivo resilience to lethal doses of bacterial lipopolysaccharide over a range of 10,000-fold. This finding suggests that proteins in serum rather than intrinsic cellular differences may play a role in regulating variations in resilience to microbe-associated molecular patterns between species. The involvement of circulating proteins as key molecules raises hope that the process might be manipulated to create better animal models and potentially new drug targets.
Article
Full-text available
Hemoglobin (Hb) uniquely associates with proinflammatory HDL in atherogenic mice and coronary heart disease (CHD) patients. In this paper, we report that Hb and its scavenger proteins, haptoglobin (Hp) and hemopexin (Hx) are significantly increased in apoA-1-containing particles of HDL both in mouse models of hyperlipidemia and in CHD patients, when compared with wild type mice and healthy donors, respectively. We further demonstrate that the association of Hb, Hp, and Hx proteins with HDL positively correlates with inflammatory properties of HDL and systemic inflammation in CHD patients. Interestingly, HDL from Hp−/− mice under atherogenic conditions does not accumulate Hb and is anti-inflammatory, suggesting that (i) Hp is required for the association of Hb with HDL and (ii) Hb·Hp complexes regulate the inflammatory properties of HDL. Moreover, treatment of apoE−/− mice with an apoA-1 mimetic peptide resulted in significant dissociation of Hb·Hp complexes from HDL and improvement of HDL inflammatory properties. Our data strongly suggest that HDL can become proinflammatory via the Hb·Hp pathway in mice and humans, and dissociation of Hb·Hp·Hx complexes from apoA-1-containing particles of HDL may be a novel target for the treatment of CHD.
Article
Full-text available
The heme-binding protein hemopexin limits TLR4 and TLR2 agonist-induced macrophage cytokine production through a mechanism distinct from heme oxygenase-1. Detection of LPS in tissues is an integral component of innate immunity that acts to protect against invasion by Gram-negative bacteria. Plasma down-regulates LPS-induced cytokine production from macrophages, thereby limiting systemic inflammation in blood and distant tissues. To identify the protein(s) involved in this process, we used classical biochemical chromatographic techniques to identify fractions of mouse sera that suppress LPS-induced TNF from bone marrow-derived macrophages (BMDMs). Fractionation yielded microgram quantities of a protein that was identified by MS to be hemopexin (Hx). Mouse Hx purified on hemin-agarose beads and rhHx decreased the production of cytokines from BMDMs and peritoneal macrophages induced by LPS. Preincubation of LPS with Hx did not affect the activity of LPS on LAL, whereas preincubation of Hx with macrophages followed by washing resulted in decreased activity of these cells in response to LPS, suggesting that Hx acts on macrophages rather than LPS. Heme-free Hx did not stimulate HO-1 in the macrophages. Purified Hx also decreased TNF and IL-6 from macrophages induced by the synthetic TLR2 agonist Pam3Cys. Our data suggest that Hx, which is an acute-phase protein that increases during inflammation, limits TLR4 and TLR2 agonist-induced macrophage cytokine production directly through a mechanism distinct from HO-1.
Article
Full-text available
Depending upon which TLRs are triggered, dendritic cells (DCs) may orient the differentiation of naive CD4(+) T cells toward either Th1, Th2, regulatory T cells, or the recently defined Th17 lineage. In this study, we report that a dual stimulation of TLR4 and TLR7/8 with LPS plus R848 leads human monocyte-derived DCs (MoDCs) to produce multiple pro- and anti-inflammatory cytokines, including IL-10, IL-12, and IL-23. Surprisingly, a significant variability in the up-regulation of these cytokines is observed in DCs obtained from various healthy donors, with approximately one of three being "high responders." High responding MoDCs stimulated via TLR4 and TLR7/8 induce naive allogeneic CD4(+) T cell to secrete sequentially IL-10 and IFN-gamma, and eventually IL-17A, whereas low responding MoDCs only stimulate IFN-gamma production. Both TLR7 and TLR8 play a central role in this phenomenon: TLR4 triggering with LPS up-regulates TLR7 expression on human MoDCs from high responders, silencing of either TLR7 or TLR8 mRNAs inhibits cytokine production in LPS plus R848-treated MoDCs, and plasmacytoid DCs constitutively expressing high levels of TLR7 induce the production of IL-10, IFN-gamma, and IL-17A by naive T cells when stimulated with R848 alone. Collectively, our results illustrate the synergy between TLR4 and TLR7/8 in controlling the sequential production of regulatory and proinflammatory cytokines by naive CD4(+) T cells. The observed polymorphism in DC responses to such TLR-mediated stimuli could explain differences in the susceptibility to infectious pathogens or autoimmune diseases within the human population.
Article
Toll-like receptors (TLRs) recognize specific molecular patterns of pathogenic microorganisms, including bacteria, fungi, protozoa, and virus. Stimulation of TLRs triggers gene expression involved in innate immune response and further instructs development of antigen-specific adaptive immunity. Molecular mechanisms by which TLRs activate innate immunity are now being elucidated through analysis of TLR-mediated signaling pathways. TLR signaling originates from the cytoplasmic Toll/IL-1 receptor (TIR) domain, which is conserved among all TLRs. In addition, recent evidence indicates that TIR domain-containing adaptors, such as MyD88, TRIF, TIRAP, and TRAM, play essential roles in TLR signaling. MyD88 is essential for inflammatory cytokine production via all TLRs, whereas TRIF mediates a MyD88-independent induction of type I IFNs via TLR3 and TLR4. TIRAP is specifically involved in TLR2-, and TLR4-mediated MyD88-dependent pathway, and TRAM acts in the TLR4-mediated TRIF-dependent pathway. Therefore, the specific functions of individual TLRs can be elicited by utilizing different combinations of TIR domain-containing adaptors. These recent progresses have made us aware of the fact that innate immunity possesses a skillful system to detect microbial invasion in the host and trigger appropriate immune responses.
Article
In late March 2009, an outbreak of a respiratory illness later proved to be caused by novel swine-origin influenza A (H1N1) virus (S-OIV) was identified in Mexico. We describe the clinical and epidemiologic characteristics of persons hospitalized for pneumonia at the national tertiary hospital for respiratory illnesses in Mexico City who had laboratory-confirmed S-OIV infection, also known as swine flu. We used retrospective medical chart reviews to collect data on the hospitalized patients. S-OIV infection was confirmed in specimens with the use of a real-time reverse-transcriptase-polymerase-chain-reaction assay. From March 24 through April 24, 2009, a total of 18 cases of pneumonia and confirmed S-OIV infection were identified among 98 patients hospitalized for acute respiratory illness at the National Institute of Respiratory Diseases in Mexico City. More than half of the 18 case patients were between 13 and 47 years of age, and only 8 had preexisting medical conditions. For 16 of the 18 patients, this was the first hospitalization for their illness; the other 2 patients were referred from other hospitals. All patients had fever, cough, dyspnea or respiratory distress, increased serum lactate dehydrogenase levels, and bilateral patchy pneumonia. Other common findings were an increased creatine kinase level (in 62% of patients) and lymphopenia (in 61%). Twelve patients required mechanical ventilation, and seven died. Within 7 days after contact with the initial case patients, a mild or moderate influenza-like illness developed in 22 health care workers; they were treated with oseltamivir, and none were hospitalized. S-OIV infection can cause severe illness, the acute respiratory distress syndrome, and death in previously healthy persons who are young to middle-aged. None of the secondary infections among health care workers were severe.
Article
Streptococcus suis, a major swine pathogen world-wide, can trigger macrophages to secrete large amounts of proinflammatory cytokines, which increase the permeability of the blood-brain barrier. In this study, we hypothesized that hemoglobin may potentiate the inflammatory response of human macrophages stimulated with a S. suis cell-wall preparation. Monocyte-derived macrophages were stimulated with the S. suis cell-wall preparation in the presence or absence of human hemoglobin, and the secretion of interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha), IL-6, and IL-8 was analyzed by enzyme-linked immunosorbent assays. The cell-wall preparation induced dose-dependent IL-1beta, TNF-alpha, IL-6, and IL-8 responses in macrophages. Hemoglobin potentiated the cell-wall induced inflammatory response, resulting in a significantly higher secretion of all the cytokines. The S. suis cell-wall preparation in combination with hemoglobin activated macrophage intracellular kinases involved in inflammatory signaling pathways. In conclusion, hemoglobin, which may be released in vivo by the action of S. suis suilysin on red blood cells, contributes to raising the levels of pro-inflammatory mediators by acting in synergy with S. suis cell-wall components. This phenomenon may contribute to the development and the severity of meningitis.