ArticlePDF AvailableLiterature Review

The Haptoglobin-CD163-Heme Oxygenase-1 Pathway for Hemoglobin Scavenging

Hindawi
Oxidative Medicine and Cellular Longevity
Authors:

Abstract and Figures

The haptoglobin- (Hp-) CD163-heme oxygenase-1 (HO-1) pathway is an efficient captor-receptor-enzyme system to circumvent the hemoglobin (Hb)/heme-induced toxicity during physiological and pathological hemolyses. In this pathway, Hb tightly binds to Hp leading to CD163-mediated uptake of the complex in macrophages followed by lysosomal Hp-Hb breakdown and HO-1-catalyzed conversion of heme into the metabolites carbon monoxide (CO), biliverdin, and iron. The plasma concentration of Hp is a limiting factor as evident during accelerated hemolysis, where the Hp depletion may cause serious Hb-induced toxicity and put pressure on backup protecting systems such as the hemopexin-CD91-HO pathway. The Hp-CD163-HO-1 pathway proteins are regulated by the acute phase mediator interleukin-6 (IL-6), but other regulatory factors indicate that this upregulation is a counteracting anti-inflammatory response during inflammation. The heme metabolites including bilirubin converted from biliverdin have overall an anti-inflammatory effect and thus reinforce the anti-inflammatory efficacy of the Hp-CD163-HO-1 pathway. Future studies of animal models of inflammation should further define the importance of the pathway in the anti-inflammatory response.
This content is subject to copyright. Terms and conditions apply.
Hindawi Publishing Corporation
Oxidative Medicine and Cellular Longevity
Volume , Article ID ,  pages
http://dx.doi.org/.//
Review Article
The Haptoglobin-CD163-Heme Oxygenase-1 Pathway for
Hemoglobin Scavenging
Jens Haugbølle Thomsen, Anders Etzerodt, Pia Svendsen, and Søren K. Moestrup
Department of Biomedicine, University of Aarhus, Ole Worms Alle 3, Building 1170, 8000 Aarhus C, Denmark
Correspondence should be addressed to Søren K. Moestrup; skm@biokemi.au.dk
Received  March ; Revised May ; Accepted  May 
Academic Editor: Mohammad Abdollahi
Copyright ©  Jens Haugbølle omsen et al. is is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium,provided the original work is properly cited.
e haptoglobin- (Hp-) CD-heme oxygenase- (HO-) pathway is an ecient captor-receptor-enzyme system to circumvent the
hemoglobin (Hb)/heme-induced toxicity during physiological and pathological hemolyses. In this pathway, Hb tightly binds to Hp
leading to CD-mediated uptake of the complex in macrophages followed by lysosomal Hp-Hb breakdown and HO--catalyzed
conversion of heme into the metabolites carbon monoxide (CO), biliverdin, and iron. e plasma concentration of Hp is a limiting
factor as evident during accelerated hemolysis, where the Hp depletion may cause serious Hb-induced toxicity and put pressure
on backup protecting systems such as the hemopexin-CD-HO pathway. e Hp-CD-HO- pathway proteins are regulated by
the acute phase mediator interleukin- (IL-), but other regulatory factors indicate that this upregulation is a counteracting anti-
inammatory response during inammation. e heme metabolites including bilirubin converted from biliverdin have overall
an anti-inammatory eect and thus reinforce the anti-inammatory ecacy of the Hp-CD-HO- pathway. Future studies of
animal models of inammation should further dene the importance of the pathway in the anti-inammatory response.
1. Introduction
Erythrocytes produced in the bone marrow have a life span of
average  days []. At this time the senescent erythrocytes
have undergone changes in the phospholipid composition in
the plasma membrane and they are recognized and phagocy-
tosed by macrophages particularly in the spleen red pulp and
the bone marrow []. Alternatively, the erythrocytes can rup-
ture in the circulation before their expected recognition by
the macrophages. is intravascular hemolysis accounts for
aboutpercentofthetotalturnoveroferythrocytesdur-
ing normal physiological conditions. Several diseases such
as hemoglobinopathies, autoimmune disorders, malaria, and
other infections may highly increase intravascular hemolysis
thus challenging the inherent Hb scavenging from plasma [].
Hbcanalsobereleasedoutsidethevascularsystemincaseof
internal bleedings such as microvascular and macrovascular
hemorrhage. e Hb release due to intravascular hemolysis
and internal bleeding may have damaging eect on the tissues
[].
e Hb-binding Hp represents a rst defense line that
instantly reduces the Hb toxicity and facilitates its removal
by CD (Figure ). is leads to proteolytic degradation of
Hb and catabolism of the toxic heme moiety via the HO-
pathway. In this review, we describe the proteins in this
pathway and their suggested role in the anti-inammatory
response.
2. Hp—Expression, Structure, and
BindingofHb
Hp is an abundant plasma glycoprotein(.–. g/L) secreted
primarily by hepatocytes but also by other cell types, such
as monocytes/macrophages and neutrophils []. It is post-
translationally cleaved into an 𝛼-anda𝛽-chain forming
a complement control protein (CCP) domain and a serine
proteinase domain, respectively []. e two domains remain
connected through disulde bridges. Furthermore, the CCP
𝛼-chain connects to another 𝛼-chain leading to the Hp 𝛼𝛽
formation, which is the basic form seen in all species. Higher
polymericformsarealsoseeninhumansbecauseofthetwo
allelic Hp variants genes Hp, and Hp [].eygiveriseto
three possible phenotypes: Hp-, Hp- and Hp-. e Hp
gene contains a duplication of a part of the Hp gene, which
Oxidative Medicine and Cellular Longevity
Haptoglobin
Hemoglobin
CD163
CD91
Hemopexin
Heme
Macrophage membrane
Cytoplasm
Receptor
recycling Ligand
endocytosis
Ligand
endocytosis
Receptor
recycling
F : e CD pathway for uptake of Hb-Hp complexes and the CD pathway for uptake of hemopexin- (Hx-) heme complexes. e
endocytosis of the ligand leads to degradation in lysosomes while the receptor recycles from the endosomes back to the plasma membrane.
results in a Hp protein with a duplicated 𝛼-chain. is causes
the formation of a range of polymeric forms of the Hp-
and Hp- phenotypes []. e phenotype is associated with
slight dierences in average plasma Hp levels (Hp- >Hp-
>Hp-) [].
Hp has dierent expression patterns in mammals and
responds to various degrees on inammation. In humans,
Hp is moderately upregulated during acute phase conditions,
where the acute phase mediators interleukin- (IL-) and IL-
further stimulate Hp synthesis in hepatocytes [,]. At sites
of inammation, Hp may be upregulated locally by release
from stored granules in activated neutrophils [].
Recent determination of the crystal structure of the
porcine Hb-Hp complex revealed a barbell-like structure
with oxygenated Hb bound to the serine proteinase domain
(𝛽-chain) of Hp. In this structure, the CCP domains were
connected by the formation of a not previously described
CCP fusion domain formed by 𝛽-strand swapping []. e
binding interface encompasses several of the amino acids
prone to oxidation in the absence of Hp, thus providing
a structural basis for the direct protective function of Hp
[]. e “loop region of the serine proteinase domain
previouslyshowntobeinvolvedinthebindingofHpto
CD protrudes from the complex [].
Hb released into plasma from ruptured erythrocytes
dissociates into dimers that instantly bind to Hp by a virtually
irreversible interaction []. It thereby directly detoxies the
oxidative Hb, prevents its ltration in the kidney [,], and
promotes the CD-mediated uptake of Hb in macrophages
[].
Hb’s toxicity relates in particular to the formation of
oxygen radicals and the scavenging of nitric oxide (NO) [].
e iron coordinated in Hb and heme reacts strongly in the
presence of hydrogen peroxide producing hydroxyl radicals
and downstream oxidation products. While sequestered in
the erythrocytes, cytoplasmic enzymes such as catalase and
superoxide dismutase limit the concentration of hydrogen
peroxide and oxygen anions and thus the oxidative reactivity
Oxidative Medicine and Cellular Longevity
ofHb.WhenboundtoHp,theoxidativeintermediateHb-
FeIV is stabilized and rendered less kinetically active [].
Additionally, Hp protects Hb from oxidative modications
that would otherwise prevent its clearance or result in release
of free heme to the circulation [,]. Binding of NO to Hb
in plasma impairs NO signaling, which may aect platelet
aggregation and increase vascular contraction []. ese are
serioussymptomsindiseaseswithstronghemolyticcrises
such as sickle cell anemia. Hp has not yet been shown to
protect against NO scavenging directly, but the observed
protection provided by Hp against nitric oxide scavenging is
probablyduetotheacceleratedclearanceofHbasmediated
by Hp [].
e binding of Hb to Hp- leads to the formation of
an approximate  kDa complex. Much larger complexes
are formed, when Hb binds to the Hp- and Hp- forms.
Whatever kind of Hb-Hp complex is formed, the complex
formation eectively reduces renal ltration of Hb [,]. In
addition, it elicits a high anity site for CD recognition
leading to clearance of Hp and Hb []. As a consequence,
hemolysis leads to consumption of Hp that can be vir-
tuallyabsent,ifthereleaseofHbintoplasmaoverrides
the production of the Hp. A low Hp level in plasma is
therefore a strong and well-known biomarker for accelerated
intravascular hemolysis. Despite circulating Hp in its free
none-Hb-bound form does not bind to CD, the Hb-bound
Hp is directly involved in the binding to CD. Extensive
mutagenesis studies of Hp have identied basic residues in Hp
loop as important residues involved in the receptor binding
[,]. It is yet not known if Hb binding is involved in CD
binding of the Hp-Hb complex.
3. Other Roles of Hp
Besides its established eect in protecting against the toxic
eectofHb,otherfunctionsofHphavebeenreported.
ese functions, which are yet less explored than the Hb-
relatedfunctionofHp,includepromotedangiogenesisandan
overall anti-inammatory eect as reviewed elsewhere [,].
Furthermore, speculations of other roles of Hp are nourished
by intriguing correlations between various diseases and Hp
phenotypes (reviewed by Levy et al. []). In diabetic patients,
the risk of cardiovascular disease is reported signicantly
higher for patients with the Hp- genotype []. In the same
group of patients, vitamin E supplementation has been shown
to be protective against these cardiovascular complications
[]. Studies of cases of subarachnoid hemorrhage also
indicate an increased risk of cerebral vasospasms in Hp-
individuals compared to Hp-. On the other hand, Hp-
has been proposed to have a protective function against
malaria [,]. A recent study demonstrates a link between
hemolysis-induced activation of the HO- and neutrophil
dysfunction which may be aected by the Hp concentration
and phenotype []. However, it should be noted that other
malaria studies have not conrmed signicant association
of Hp genotype on disease outcome []. Further epi-
demiological analyses and biochemical studies are warranted
to document and mechanistically understand associations
between Hp phenotype and disease.
4. CD163—Structure, Expression, and
Receptor Function for Hp-Hb
CD is a  kDa transmembrane glycoprotein expressed
exclusively in cells of the reticuloendothelial system. It is a
member of the “scavenger receptor cysteine rich (SRCR)
superfamily class B. is receptor family is characterized by
containing one or more SRCR domains that are conserved
domains consisting of – amino acids and to cysteine
residues connected by disulde bridges []. Crystallization
of the repeat in other proteins has revealed a compact fold
of - 𝛽-sheets cradling an 𝛼-helix []. Class A and
classBSRCRdomainsarestructurallysimilarwithonlya
few slight dierences. Class B domains are translated from
a single exon, and class A domains are from two exons and
they contain one more disulde bridge than class B []. e
extracellular segment of CD contains nine SRCR domains
only separated by a  proline/serine/threonine-rich linker
region between domain and [].
Four dierent isoforms have been demonstrated, result-
ing from alternative splicing of the RNA encoding the
cytoplasmic tail []. e shortest and most abundant variant
consists of  amino acid residues, while the longest consists
ofand,respectively.erstaminoacidsaer
the transmembrane segment are identical amongst the iso-
forms and contain phosphorylation motives for casein kinase
and protein kinase C []. Other possible phosphorylation
motives are present on the longer isoforms []. Confocal
microscopy has revealed that the shortest tail variant is pri-
marily present in the cell membrane while the longer variants
are located in the endosomal/Golgi cellular compartment
[].
CD is expressed exclusively on cells of the monocyte-
macrophage cell lineage. A high expression is seen in most
mature tissue macrophages such as Kuper cells in the liver,
red pulp macrophages in the spleen, resident bone marrow
macrophages, and alveolar macrophages in the lungs [].
Cell types derived from monocytes showing low or no CD
expression include dendritic cells, Langerhans cells, and
white pulp macrophages in the spleen [,].
Several endogenous and exogenous molecules have been
shown to regulate the expression of CD in in vitro
experiments. Glucocorticoids, IL-, and interleukin- (IL-
) strongly upregulate its expression, whereas interferon-
𝛾(IFN𝛾), tumor necrosis factor-𝛼(TNF-𝛼), interleukin-
(IL-), granulocyte/macrophage colony stimulating factor
(GM-CSF), lipopolysaccharide (LPS), and CXC-chemokine
ligand (CXCL) downregulate CD expression [,,
]. e upregulation of CD by glucocorticoids has also
been demonstrated in human volunteers following injection
with the glucocorticoid prednylidene []. Whereas IL-
has both pro- and anti-inammatory eects [], the
overall pattern is that CD expression is induced by anti-
inammatory mediators and reduced by proinammatory
molecules.
Experimental studies have shown that CD is expressed
on macrophages matching the phenotype dened by in
vitro dierentiation in response to IL- and interleukin-
(IL-) (M/alternatively activated macrophages) despite the
Oxidative Medicine and Cellular Longevity
fact that IL- alone decreases CD expression in mono-
cyte/macrophages [,]. CD positive macrophages of a
similar phenotype are abundant in the resolution phase of the
inammatory process []. ese ndings have been used to
hypothesize that CD is a marker of an anti-inammatory
and tissue homeostatic macrophage subclass []. CD is
now widely used as a marker for the macrophage class.
Finally, a novel macrophage subtype designated Mhem is
dened by a high CD and a low mannose receptor
expression []. ese macrophages have been described in
atherosclerotic lesions and they were suggested to exhibit an
antiatherogenic phenotype when examined in vivo [].
A soluble form of CD (sCD) is present in plasma
anditisupregulatedinanumberofdiseasesinvolving
macrophagesasrecentlyreviewedbyMoller[]. It is
generated by ectodomain shedding of the extracellular part of
the receptor. Both TNF-𝛼cleaving enzyme (TACE)/ADAM
and neutrophil elastase have been reported as enzymes
responsible for the cleavage []. However, the concomitant
increase in sCD and TNF-𝛼in humans exposed to LPS
does suggest an important role of TACE/ADAM, which
is activated by LPS in macrophages []. e biological
function of sCD is not yet clear, although several possible
functions have been proposed—including opsonization of
Staph. Aureus [], inhibition of T-cell proliferation []
and inhibition of tumor necrosis factor-like weak inducer of
apoptosis (TWEAK) [].
e third SRCR domain of CD is involved in the
Ca2+-dependent binding of the Hp-Hb complex [,].
e subsequent endocytosis of the ligand bound receptor
is dependent on the endocytic motifs in the cytoplasmic
tail [,]. e various CD isoforms dier in endocytic
ecacy with the shortest variant demonstrating the fastest
uptake [].
In addition to its uptake of the Hb-Hp complex CD
can facilitate the uptake of free Hb. is allows CD to
act as its own fail-safe system in pathological situations
where Hp is depleted due to excessive intravascular hemolysis
[].Towhatdegreethisfunctionisimplicatedinhuman
disease is unknown. Studies in the mouse Hp-Hb system
have disclosed subtle dierences. In this species, Hb binds
with higher anity to CD and the binding of Hp to Hb
does not further increase anity for CD []. e CD-
mediated uptake of Hb (in complex with Hp or not) induces
the secretion of IL- and IL-, as well as it upregulates several
genes responsible for the degradation of Hb-including HO-
[,].
5. Other Potential Functions of CD163
Several functions besides the scavenging of Hb have been
proposed for CD. In rats, CD expressed on resident
bone marrow macrophages has been shown to bind erythrob-
lasts and promote growth and/or survival in erythropoiesis
[]. A recent study indicates a role of CD as a pathogen-
associated molecular pattern (PAMP) receptor []. CD
demonstrated binding to both gram-positive and negative
bacteria and the bacteria induced TNF-𝛼secretion from
human monocytes []. TWEAK has been shown to be
bound and internalized by CD, indicating CD as a
possible regulator of this cytokine []—in addition to the
regulation of TWEAK by sCD as mentioned earlier. A
high sCD and low TWEAK concentration has been shown
to correlate with intima-media thickness, cardiovascular
mortality in peripheral arterial disease, and a type diabetes
diagnosis [,,]. Finally, porcine CD has been
implicated in the entry mechanisms of African swine fever
virus (ASFV) and the porcine reproductive and respiratory
syndrome virus (PRRSV) infecting myeloid cells [,].
6. Physiological Back-Up Systems
for the Heme-Protective Function of
the Hp-Hb Pathway
Excessive hemolysis as seen during malaria, sickle cell ane-
mia, autoimmune hemolysis, and many other conditions with
pathological intravascular hemolysis may lead to depletion of
Hp in plasma []. In such cases, Hb accumulates in plasma
with toxic consequences. Hb may then be taken up directly
by CD by a yet unknown pathway, be ltered in the kidney
or be degraded in plasma. e absence of Hp binding to Hb
leads to release of heme that then binds to heme-binding
proteins such as albumin, 𝛼-microglobin, and hemopexin.
Hemopexin binds heme with the highest anity leading to
uptake via LDL-receptor related protein (LRP) []/CD
(Figure ), an abundant receptor in macrophages, hepatocytes
and other cells []. Studies of hemopexin-decient mice
with and without a Hp gene knockout background have
evidenced that hemopexin constitutes a backup system for
the heme-protective role of Hp [].
7. HO-1
Hb internalized through interaction with CD is trans-
ferred to early endosomes and subsequently degraded to
heme, bioactive peptides, and amino acids [,]. HO
is responsible for the further enzymatic heme catabolism
resulting in the degradation products carbonmonoxide (CO),
ferrous iron (Fe2+) and biliverdin. Biliverdin is reduced to
bilirubin by the biliverdin reductase (Figure ).
ree isoforms of HO diering in tissue distribution,
regulation and proposed function have been identied.
HO- ( kDa) is expressed in many cell types includ-
ing macrophages. It is highly inducible in response to a
wide range of factors []. HO- ( kDa) is constitutively
expressed with the highest expression in testis and brain
[]. Finally, an HO- isoform was identied in rats but later
studies suggest it may be a pseudogene with no apparent
function [,].
HO- is a monomeric enzyme anchored to the outer
membrane of a microsomal membrane by a hydrophobic C-
terminal domain []. More recently HO- has also been
identied in caveolae demonstrating direct interaction with
caveolin- []. Whether the heme oxidation takes place in
the cytosol or in endosomal vesicles is not fully outlined
[]. Proteolytic cleavage of the active site of HO- from
the membrane-anchored C-tail occurs following hypoxia or
Oxidative Medicine and Cellular Longevity
CO
Biliverdin
Heme
Lysosome Bilirubin
CRP Fe2+
HO-1
F : e intracellular pathway for heme-degradation subse-
quent to CD and CD mediated endocytosis in macrophages.
Free heme is degraded to biliverdin, CO, and ferrous iron by the
endoplasmic reticulum enzyme HO- facing the cytosol. Electrons
are delivered by NADPH p cytochrome reductase. Biliverdin is
reduced to bilirubin by biliverdin reductase and transported to the
liver bound to albumin.
heme loading leading to translocalization of the truncated
enzyme to the nucleus. Here it promotes transcription of
antioxidative related genes including activating protein-
(AP-) []. e binding site for heme is located between
two concave 𝛼-helixes termed the proximal and distal helix,
respectively [].
e enzymatic process leading to the degradation of heme
comprises three major steps. In the rst step heme is oxidized
to hydroxyheme, and in the second step verdoheme is formed
and CO is released. e third step results in biliverdin and
Fe2+ []. e last step is rate limiting but is also the least
characterized []. During the process three molecules of
oxygen and seven electrons are consumed []. e electron
donor is NADPH cytochrome p reductase, which is
anchored alongside HO- on the endoplasmic reticulum on
the side facing the cytosol [].
e molecular mechanisms for the regulation of HO-
have been extensively investigated and the complexities
of the pre-translational regulation are now steadily being
unraveled although hampered by major dierences between
the examined species and between cell types [,]. e
expressionofHO-isinduciblebyalonglistofendogenous
and exogenous molecules []. In the context of this review,
it should be noted that heme itself aside from functioning
as cofactor and substrate of HO- also seems as the most
potent inducer of HO- expression. Other inducers include,
but are far from being limited to, heat, ultraviolet radiation,
LPS, hydrogen peroxide, several dietary phytochemical, IL-
𝛼,TNF𝛼,andNO[]. Interestingly, IL-, which
has a central role in the CD regulation, also stimulates
synthesis of HO- []. Some common mechanisms have
been proposed based upon shared cellular eects of some the
inducers: a transient increase in intracellular heme, increased
production of reactive oxygen species (ROS) generation, and
glutathione depletion [].
Many studies have demonstrated a role for protein phos-
phorylation dependent signaling pathways in the observed
HO- upregulation. A growing body of evidence points to
a central role for the mitogen associated protein kinases
(MAPK) family of kinases in this. MAPK proteins belong to
the serine/threonine kinase superfamily and is involved in
mediating signals for cell growth, dierentiation, and apop-
tosis and commonly activated in response to stressors [].
PI K/Akt, protein kinase A, protein kinase C, and tyrosine
kinase have also been implicated as possible mediators of
HO- induction [].
e existence of multiple HO- inducers corresponds
with the abundance of response elements and cis-acting
elements in the promoter region of Hmox- (the human HO-
gene). e promoter region spans at least  kb from the
󸀠start of Hmox- and contains several consensus binding
motives for binding of transcription factors such as ARE
(antioxidant response element or stress-related response
element) which is found in the promoters of proteins asso-
ciated with anti-oxidative functions (also known as phase II
enzymes) []. Its ligands include transcriptions factors
of the basic leucine zipper-superfamily of which several have
been shown to induce HO- transcription. NF-E related
factor (Nrf-) belongs to this family as well and a growing
body of evidence shows that it is essential for ARE-binding
and HO- induction [,].
Until recently, no direct molecular link between increased
oxidative stress and transcriptional activity was known.
e identication of the interactions between Nrf- and
Kelch-like ECH associated protein- (Keap-) has provided
such a link. Under low-stress conditions Keap- binds Nrf-
in the cytoplasm and directs it to ubiquitin-dependent
proteasomal degradation [,]. Oxidation of specic
cysteine residues in Keap- or the phosphorylation of Nrf-
inhibits its degradation and result in nuclear translocation,
heterodimerization,and transcriptional activity of Nrf- [].
is model explains both the observed link between MAPK
activation and Nrf- transcriptional activity [,]and
the link between oxidative stress/ROS and HO- expression.
Using knock-out technology and genetic transduction a pro-
tectiveroleofHO-inavarietyofdiseasemodelsincluding
atherosclerosis, hypoxia, hyperoxia-induced lung damage,
liver failure, liver allogra, hypertension and reperfusion
injury has been shown [].
8. The Physiological Effects of
Heme Metabolites
CO is most widely known as a toxic inhaled gas inhibiting
oxygen binding and release from Hb thus causing asphyxia-
tion. However, as mentioned earlier, it is also endogenously
produced during heme oxidation by HO-. At these relative
low levels an increasing body of evidence indicates that
CO serves several benecial physiological functions. Most
of these cellular eects are believed to be mediated by CO
binding to heme in heme-proteins []. Of notice, CO binds
andactivatessolubleguanylatecyclase(sGC)toproduce
cyclic guanosine monophosphate similarly to nitric oxide
Oxidative Medicine and Cellular Longevity
though with less ecacy []. is may mimic nitric oxide’s
well-established cytoprotective eects and this is believed
to be a major contributor to the observed cytoprotection
mediated by HO-/CO [,].
CO has also been shown to cause vasodilatation via sGC
independent activation of potassium channels in vascular
smooth muscle cells []. Furthermore, CO is believed
to modulate p MAPK in an sGC independent way
by inhibiting the expression of classical pro-inammatory
cytokines such as TNF-𝛼,IL-𝛽,andmacrophageinamma-
tory protein-𝛽while promoting the expression of the anti-
inammatory cytokine IL- []. ese anti-inammatory
eects have been demonstrated in vivo where administered
CO in nonlethal concentrations was able to reduce the
inammation induced by mechanical ventilation [,].
Cytochromecoxidase(COX)isaheme-proteininthe
inner mitochondrial membrane which transfers protons and
electrons to O2, creating water and providing energy for
transport of two protons across the membrane. CO binds
and inhibits this protein thus inhibiting O2consumption
and stimulating ROS from the accumulating electron carriers
in the mitochondria. is process is termed mitochondrial
redox signaling, and it is believed to stimulate mitochondrial
biogenesis and angiogenesis [].
e CO cleaved from hydroxyheme is primarily removed
from the body via respiration. CO diuses readily cross-
cell membranes and binds Hb with approximately -fold
higher anity than O2. Provided adequate circulation and
respiration, it is then transported to the alveoli and diuses to
the alveolar gas. Additionally, CO is slowly oxidized by COX
to CO2[]. e exhalation of CO can be used as a measure
of heme catabolism in the body [].
Iron (Fe2+)isreleasedfromhemeduringthelastenzy-
matic step of its conversion to biliverdin. A P-type ATPase
iron transporter is colocalized with HO- in the microsomal
membrane []. e importance of this iron release is
suggested because of the anemia and iron accumulation in
theliverandkidneyinHO-knock-outmiceandintherst
reported case of human HO- deciency [,,]. Most
likely, the iron released from heme enters a labile pool of
intracellular iron, available for cellular processes involving
iron or cellular export via the hepcidin-regulated ferroportin
protein in the membrane.
Intracellular iron is oxidized and bound to the ubiquitous
apoprotein ferritin. An increase in the intracellular iron
deposit aects the posttranscriptional expression of several
proteins by interaction with iron regulatory proteins and
mRNA iron response elements []. Via this mechanism
ferritin is upregulated by increased HO- activity [].
Ferritin has been shown to have antiapoptotic eects and
provide cytoprotection against oxidative damage [,].
Iron is exported from the cell by ferroportin and transported
bound to the plasma protein transferrin. Transferrin-bound
iron is taken up by cells expressing the transferrin receptor
and recycled.
e main product of heme degradation, biliverdin, is a
greenish water-soluble pigment. It is reduced to bilirubin by
biliverdin reductase. Bilirubin is a hydrophobic, yellowish
T : Major reportedc ytoprotective andantiinammatory eects
of the Hp-CD-HO- pathway.
Intravascular HpHb complex formation:
Protects against oxidative “hot spot” in Hb
Protects against heme release from Hb
Facilitates CD-mediated clearance
Prevents renal ltration of Hb and uptake in proximal tubules
Prevents NO scavenging
Cellular response on CD-mediated Hb endocytosis:
Cellular dierentiation
HO- upregulation
Nrf- activation
IL- synthesis
Other eects of heme metabolites generated by HO- activity:
Antagonism of proinammatory cytokines
ROS scavenging
Angiogenesis
Inhibition of platelet aggregation
Vaso d i l a t a t i o n
pigmentandistransportedintheplasmaboundtoalbumin.
Intheliver,bilirubinisconjugatedandexcretedinthebile.
For decades bilirubin has been considered a toxic byprod-
uct of heme degradation. Recent studies have, however,
also demonstrated potential benecial functions of bilirubin
and biliverdin in the circulation and extravascular tissues
[]. Epidemiological studies have revealed that moderately
increased plasma levels of bilirubin decrease the risk of
developing cardiovascular diseases []. In vitro studies
have demonstrated bilirubin and biliverdin as functional
antioxidants []. Biliverdin reductase has also been shown
to be at least partly responsible for HO--mediated anti-
oxidative protection []. Additionally, biliverdin reduc-
tase has been shown to promote an anti-inammatory
response in macrophages through transcriptional regulation
[].
9. Perspectives
e Hp-CD-HO- pathway for degradation of
hemoglobin is as an important and apparently to some extent
a coordinately regulated pathway that by direct hemoglobin
binding and subsequent clearance from plasma prevents
toxic and proinammatory eects of heme and hemoglobin.
In addition, the proteins in the pathway and the metabolic
heme products reinforce an anti-inammatory response.
Table summarizes major anti-inammatory eects of this
pathway. Future studies of various inammatory conditions
in vitro and in vivo models should further delineate the
molecular mechanism and elucidate if the proteins of the
pathway have anti-inammatory eects independent of
heme.Finally,thispathwayseemsasapotentialtargetfor
stimulation of the inammatory response by small molecule
drugs.
Oxidative Medicine and Cellular Longevity
Acknowledgment
is study was supported by ERC to the project TROJA.
References
[] D. Bratosin, J. Mazurier, J. P. Tissier et al., “Cellular and
molecular mechanisms of senescent erythrocyte phagocytosis
by macrophages. A review, Biochimie,vol.,no.,pp.
, .
[]M.Straat,R.vanBruggen,D.deKorte,andN.P.Juer-
mans, “Red blood cell clearance in inammation, Trans f usio n
Medicine and Hemotherapy,vol.,no.,pp.,.
[] R. P. Rother, L. Bell, P. Hillmen, and M. T. Gladwin, “e clinical
sequelae of intravascular hemolysis and extracellular plasma
hemoglobin: a novel mechanism of human disease, Journal of
the American Medical Association,vol.,no.,pp.
, .
[] J. Aronowski and X. Zhao, “Molecular pathophysiology of
cerebral hemorrhage: secondary brain injury, Stroke,vol.,
no. , pp. –, .
[] A. P. Levy, R. Asleh, S. Blum et al., “Haptoglobin: basic and
clinical aspects, Antioxidants and Redox Signaling,vol.,no.
, pp. –, .
[] K. eilgaard-M¨
onch, L. C. Jacobsen, M. J. Nielsen et al.,
“Haptoglobin is synthesized during granulocyte dierentiation,
stored in specic granules, and released by neutrophils in
response to activation, Blood,vol.,no.,pp.,.
[]C.B.Andersen,M.Torvund-Jensen,M.J.Nielsenetal.,
“Structure of the haptoglobin-haemoglobin complex, Nature,
vol. , no. , pp. –, .
[] K. B. Wicher and E. Fries, “Evolutionary aspects of hemoglobin
scavengers, Antioxidants and Redox Signaling,vol.,no.,pp.
–, .
[]M.J.NielsenandS.K.Moestrup,“Receptortargetingof
hemoglobin mediated by the haptoglobins: roles beyond heme
scavenging, Blood,vol.,no.,pp.,.
[] J. G. Bode, U. Albrecht, D. Haussinger, P. C. Heinrich, and F.
Schaper, “Hepatic acute phase proteins—regulation by IL- and
IL--type cytokines involving STAT and its crosstalk with NF-
kappaB-dependent signaling, European Journal of Cell Biology,
vol. , no. -, pp. –, .
[] A. I. Alayash, C. B. Andersen, S. K. Moestrup, and L. B¨
ulow,
“Haptoglobin: the hemoglobin detoxier in plasma, Trend s in
Biotechnology,vol.,no.,pp.,.
[] V.P.D.A.Wobeto,T.R.Zaccariotto,andM.D.F.Sonati,“Poly-
morphism of human haptoglobin and its clinical importance,
Genetics and Molecular Biology,vol.,no.,pp.,.
[] A. Etzerodt and S. K. Moestrup, “CD and inammation:
biological, diagnostic and therapeutic aspects, Antioxid Redox
Signal,vol.,no.,pp.,.
[]S.Banerjee,Y.Jia,C.J.P.Siburtetal.,“Haptoglobinalters
oxygenation and oxidation of hemoglobin and decreases prop-
agation of peroxide-induced oxidative reactions, Free Radical
Biology and Medicine,vol.,no.,pp.,.
[] P.W.Buehler,B.Abraham,F.Vallelianetal.,“Haptoglobinpre-
serves the CD hemoglobin scavenger pathway by shielding
hemoglobin from peroxidative modication, Blood,vol.,no.
, pp. –, .
[] E. Tolosano, S. Fagoonee, N. Morello, F. Vinchi, and V. Fiorito,
“Heme scavenging and the other facets of hemopexin, Antiox-
idants and Redox Signaling,vol.,no.,pp.,.
[] I. Azarov, X. He, A. Jeers et al., “Rate of nitric oxide scavenging
by hemoglobin bound to haptoglobin, Nitric Oxide,vol.,no.
, pp. –, .
[] M. Kristiansen, J. H. Graversen, C. Jacobsen et al., “Identica-
tion of the haemoglobin scavenger receptor,” Nature,vol.,
no.,pp.,.
[] M. J. Nielsen, S. V. Petersen, C. Jacobsen et al., A unique
loop extension in the serine protease domain of haptoglobin is
essential for CD recognition of the haptoglobin-hemoglobin
complex, JournalofBiologicalChemistry,vol.,no.,pp.
–, .
[] M. J. Nielsen, C. B. F. Andersen, and S. K. Moestrup, “CD
binding to haptoglobin-hemoglobin complexes involves a dual-
point electrostatic receptor-ligand pairing, Journal of Biological
Chemistry,.
[] A. P. Levy, I. Hochberg, K. Jablonski et al., “Haptoglobin
phenotype is an independent risk factor for cardiovascular
disease in individuals with diabetes: the strong heart study,
JournaloftheAmericanCollegeofCardiology,vol.,no.,pp.
–, .
[] A. P. Lev y, S. Blum, M. Vardi et al., “Vitamin e reduces
cardiovascular disease in individuals with diabetes mellitus and
the haptoglobin - genotype, Pharmacogenomics,vol.,no.,
pp. –, .
[] A. A. Elagib, A. O. Kider, B. ˚
Akerstr¨
om, and M. I. Elbashir,
Association of the haptoglobin phenotype (-) with falc iparum
malaria in Sudan, Transactions of the Royal Society of Tropical
Medicine and Hygiene, vol. , no. , pp. –, .
[] I.K.Quaye,F.A.Ekuban,B.Q.Gokaetal.,“Haptoglobin-is
associated with susceptibility to severe Plasmodium falciparum
malaria, Transactions of the Royal Society of Tropical Medicine
and Hygiene,vol.,no.,pp.,.
[] A. J. Cunnington, M. Njie, S. Correa, E. N. Takem, E. M.
Riley,andM.Walther,“Prolongedneutrophildysfunctionaer
Plasmodium falciparum malaria is related to hemolysis and
heme oxygenase- induction, Journal of Immunology,vol.,
no. , pp. –, .
[] V.R.Mendonca,N.F.Luz,N.J.G.Santosetal.,“Association
between the haptoglobin and heme oxygenase genetic proles
and soluble CD in susceptibility to and severity of human
malaria, Infection and Immunity,vol.,no.,pp.,
.
[] I. Quaye, “Haptoglobin genotypes are not associated with
resistance to severe malaria in e Gambia,” Transactions of the
Royal Society of Tropical Medicine and Hygiene,vol.,no.,p.
, .
[] B.Beiguelman,F.P.Alves,M.M.Mouraetal.,“eassociation
of genetic markers and malaria infection in the Brazilian
Western Amazonian region, Memorias do Instituto Oswaldo
Cruz, vol. , no. , pp. –, .
[] M.Freeman,J.Ashkenas,D.J.G.Reesetal.,“Anancient,highly
conserved family of cysteine-rich protein domains revealed
by cloning type I and type II murine macrophage scavenger
receptors, Proceedings of the National Academy of Sciences of
the United States of America,vol.,no.,pp.,.
[] B.Rodamilans,I.G.Mu
˜
noz, E. Bragado-Nilsson et al., “Crystal
structure of the third extracellular domain of CD reveals the
fold of a group B scavenger cysteine-rich receptor domain,
Journal of Biological Chemistry,vol.,no.,pp.,
.
[] E. Hohenester, T. Sasaki, and R. Timpl, Crystal structure
of a scavenger receptor cysteine-rich domain sheds light on
Oxidative Medicine and Cellular Longevity
an ancient superfamily, Nature Structural Biology,vol.,no.,
pp. –, .
[] J. R. Somoza, J. D. Ho, C. Luong et al., “e structure of the
extracellular region of human hepsin reveals a serine protease
domain and a novel scavenger receptor cysteine-rich (SRCR)
domain, Structure, vol. , no. , pp. –, .
[] V. G. Mart´
ınez,S.K.Moestrup,U.Holmskov,J.Mollenhauer,
and F. Lozano, “e conserved scavenger receptor cysteine-
rich super family in therapy and diagnosis, Pharmacological
Reviews,vol.,no.,pp.,.
[] H. Van Gorp, P. L. Delputte, and H. J. Nauwynck, “Scavenger
receptor CD, a Jack-of-all-trades and potential target forcell-
directed therapy, Molecular Immunology,vol.,no.-,pp.
–, .
[] M.Ritter,C.Buechler,T.Langmann,E.Orso,J.Klucken,andG.
Schmitz, “e scavenger receptor CD: regulation, promoter
structure and genomic organization, Pathobiology,vol.,no.
-, pp. –, .
[] M. Ritter, C. Buechler, M. Kapinsky, and G. Schmitz, “Interac-
tion of CD with the regulatory subunit of casein kinase II
(CKII) and dependence of CD signaling on CKII and protein
kinase C, European Journal of Immunology,vol.,no.,pp.
–, .
[] M.J.Nielsen,M.Madsen,H.J.Møller,andS.K.Moestrup,“e
macrophage scavenger receptor CD: endocytic properties of
cytoplasmic tail variants, Journal of Leukocyte Biology,vol.,
no. , pp. –, .
[] M. B. Maniecki, H. J. Møller, S. K. Moestrup, and B. K. Møller,
“CD positive subsets of blood dendritic cells: the scavenging
macrophage receptors CD and CD are coexpressed on
human dendritic cells and monocytes, Immunobiology, vol. ,
no. -, pp. –, .
[] V. Kodelja and S. Goerdt, “Dissection of macrophage dier-
entiation pathways in cutaneous macrophage disorders and in
vitro, Experimental Dermatology,vol.,no.,pp.,
.
[] G. Zwadlo-Klarwasser, S. Bent, H. D. Haubeck, C. Sorg, and
W. Schmutzler, “Glucocorticoid-induced appearance of the
macrophage subtype RM / in peripheral blood of man,
International Archives of Allergy and Applied Immunology,vol.
,no.,pp.,.
[] D. Kamimura, K. Ishihara, and T. Hirano, “IL- signal transduc-
tion and its physiological roles: the signal orchestration model,
Reviews of Physiology, Biochemistry and Pharmacology,vol.,
pp. –, .
[] S. Gordon and F. O. Martinez, Alternative activation of
macrophages: mechanism and functions, Immunity,vol.,no.
, pp. –, .
[] G. Zwadlo, R. Voegeli, K. Schulze Ostho, and C. Sorg, A mon-
oclonal antibody to a novel dierentiation antigen on human
macrophages associated with the down-regulatory phase of the
inammatory process, Experimental Cell Biology,vol.,no.,
pp.,.
[] A. V. Finn, M. Nakano, R. Polavarapu et al., “Hemoglobin
directs macrophage dierentiation and prevents foam cell
formationinhumanatheroscleroticplaques,Journal of the
American College of Cardiology,vol.,no.,pp.,.
[] J. J. Boyle, “Heme and haemoglobin direct macrophage Mhem
phenotype and counter foam cell formation in areas of
intraplaque haemorrhage, Current Opinion in Lipidology,vol.
,no.,pp.,.
[] H. J. Moller, “Soluble CD, Scandinavian Journal of Clinical
& Laboratory Investigation,vol.,no.,pp.,.
[] A. Etzerodt, M. B. Maniecki, K. Møller, H. J. Møller, and S.
K.Moestrup,“Tumornecrosisfactor𝛼-converting enzyme
(TACE/ADAM) mediates ectodomain shedding of the scav-
enger receptor CD, Journal of Leukocyte Biology, vol. , no.
, pp. –, .
[] J. Kneidl, B. L¨
oer,M.C.Eratetal.,“SolubleCDpromotes
recognition, phagocytosis and killing of Staphylococcus aureus
via binding of specic bronectin peptides, Cellular Microbiol-
ogy, vol. , no. , pp. –, .
[] D. Baeten, H. J. Møller, J. Delanghe, E. M. Veys, S. K. Moestrup,
and F. De Keyser, Association of CD+ macrophages and
local production of soluble CD with decreased lympho-
cyte activation in spondylarthropathy synovitis, Arthritis and
Rheumatism,vol.,no.,pp.,.
[] J. A. Moreno, B. Mu˜
noz-Garc´
ıa,J.L.Mart
´
ın-Ventura et al.,
“e CD-expressing macrophages recognize and inter-
nalize TWEAK. Potential consequences in atherosclerosis,
Atherosclerosis,vol.,no.,pp.,.
[] M. Madsen, H. J. Møller, M. J. Nielsen et al., “Molecular char-
acterization of the haptoglobin-hemoglobin receptor CD:
ligand binding properties of the scavenger receptor cysteine-
rich domain region, JournalofBiologicalChemistry,vol.,
no. , pp. –, .
[] C. A. Schaer, G. Schoedon, A. Imhof, M. O. Kurrer, and
D. J. Schaer, “Constitutive endocytosis of CD mediates
hemoglobin-heme uptake and determines the noninamma-
tory and protective transcriptional response of macrophages to
hemoglobin, Circulation Research,vol.,no.,pp.,
.
[] D. J. Schaer, C. A. Schaer, P. W. Buehler et al., “CD is
the macrophage scavenger receptor for native and chemically
modied hemoglobins in the absence of haptoglobin, Blood,
vol. , no. , pp. –, .
[] A. Etzerodt, M. Kjolby, M. J. Nielsen, M. Maniecki, P. Svendsen,
and S. K. Moestrup, “Plasma clearance of hemoglobin and hap-
toglobin in mice and eect of CD gene targeting disruption,
Antioxidants & Redox Signaling,vol.,no.,pp.,
.
[] P. Philippidis, J. C. Mason, B. J. Evans et al., “Hemoglobin
scavenger receptor CD mediates interleukin- release
and heme oxygenase- synthesis: antiinammatory monocyte-
macrophage responses in vitro, in resolving skin blisters in
vivo, and aer cardiopulmonar y bypass surgery, Circulation
Research, vol. , no. , pp. –, .
[] B. O. Fabriek, M. M. J. Poliet, R. P. M. Vloet et al., “e
macrophage CD surface glycoprotein is an erythroblast
adhesion receptor, Blood,vol.,no.,pp.,.
[] B. O. Fabriek, R. V. Bruggen, D. M. Deng et al., “e macrophage
scavenger receptor CD functions as an innate immune
sensor for bacteria, Blood,vol.,no.,pp.,.
[] L. C. Bover, M. Card´
o-Vila, A. Kuniyasu et al., A previously
unrecognized protein-protein interaction between TWEAK
and CD: potential biological implications, Journal of
Immunology,vol.,no.,pp.,.
[] G.Llaurado,J.M.Gonz
´
alez-Clemente, E. Maym´
o-Masip, D.
Sub´
ıas, J. Vendrell, and M. R. Chac ´
on, “Serum levels of TWEAK
and scavenger receptor CD in type diabetes mellitus:
relationship with cardiovascular risk factors. a case-control
study, PLoS ONE,vol.,no.,ArticleIDe,.
Oxidative Medicine and Cellular Longevity
[] G. Urbonaviciene, J. L. Martin-Ventura, J. S. Lindholt et al.,
“Impact of soluble TWEAK and CD/TWEAK ratio on
long-termcardiovascularmortalityinpatientswithperipheral
arterial disease, Atherosclerosis,vol.,no.,pp.,
.
[] C. S´
anchez-Torres, P. G´
omez-Puertas, M. G´
omez-Del-Moral et
al., “Expression of porcine CD on monocytes/macrophages
correlates with permissiveness to African swine fever infection,
Archives of Virology,vol.,no.,pp.,.
[] S. K. W. Welch and J. G. Calvert, A brief review of CD and
its role in PRRSV infection, Virus R es earch,vol.,no.-,pp.
–, .
[] J. D. Belcher, J. D. Beckman, G. Balla, J. Balla, and G. Vercellotti,
“Heme degradation and vascular injury,” Antioxidants and
Redox Signaling,vol.,no.,pp.,.
[] V. Hvidberg, M. B. Maniecki, C. Jacobsen, P. Højrup, H. J.
Møller, and S. K. Moestrup, “Identication of the receptor
scavenging hemopexin-heme complexes, Blood,vol.,no.,
pp. –, .
[] S. K. Moestrup, J. Gliemann, and G. Pallesen, “Distribu-
tion of the 𝛼-macroglobulin receptor/low density lipoprotein
receptor-related protein in human tissues,” Cell and Tissue
Research,vol.,no.,pp.,.
[] E. Tolosano, S. Fagoonee, E. Hirsch et al., “Enhanced
splenomegaly and severe liver inammation in
haptoglobin/hemopexin double-null mice aer acute
hemolysis, Blood,vol.,no.,pp.,.
[]I.Fruitier,I.Garreau,A.Lacroix,A.Cupo,andJ.M.Piot,
“Proteolytic degradation of hemoglobin by endogenous lysoso-
mal proteases gives rise to bioactive peptides: hemorphins, e
FEBS Letters,vol.,no.,pp.,.
[] C. Y. Wang and L. Y. Chau, “Heme oxygenase- in cardiovascu-
lar diseases: molecular mechanisms and clinical perspectives,
Chang Gung Medical Journal,vol.,no.,pp.,.
[] M.L.Wu,Y.C.Ho,C.Y.Lin,andS.F.Yet,“Hemeoxygenase-
in inammation and cardiovascular disease, American Journal
of Cardiovascular Disease,vol.,no.,pp.,.
[] S. Hayashi, Y. Omata, H. Sakamoto et al., “Characterization of
rat heme oxygenase- gene. Implication of processed pseudo-
genes derived from heme oxygenase- gene, Gene,vol.,no.
, pp. –, .
[] W. K. Mccoubrey, T. J. Huang, and M. D. Maines, “Isolation
and characterization of a cDNA from the rat brain that encodes
hemoprotein heme oxygenase-, European Journal of Biochem-
istry,vol.,no.,pp.,.
[] T. Yoshida and M. Sato, “Posttranslational and direct integration
of heme oxygenase into microsomes, Biochemical and Biophys-
ical Research Communications,vol.,no.,pp.,
.
[] N. H. Jung, H. P. Kim, B. R. Kim et al., “Evidence for
heme oxygenase- association with caveolin- and - in mouse
mesangial cells, IUBMB Life,vol.,no.,pp.,.
[] C. Delaby, C. Rondeau, C. Pouzet et al., “Subcellular localization
of iron and heme metabolism related proteins at early stages of
erythrophagocytosis, PLoS ONE,vol.,no.,ArticleIDe,
.
[]Q.Lin,S.Weis,G.Yangetal.,“Hemeoxygenase-protein
localizes to the nucleus and activates transcription factors
important in oxidative stress, JournalofBiologicalChemistry,
vol. , no. , pp. –, .
[] M.Unno,T.Matsui,andM.Ikeda-Saito,“Structureandcatalytic
mechanism of heme oxygenase, Natural Product Reports,vol.
,no.,pp.,.
[] M. Unno, T. Matsui, and M. Ikeda-Saito, Crystallographic
studies of heme oxygenase complexed with an unstable reaction
intermediate, verdoheme, Journal of Inorganic Biochemistry,
vol. , pp. –, .
[] T. Matsui, A. Nakajima, H. Fujii et al., O2-andH
2O2-
dependent verdoheme degradation by heme oxygenase: reac-
tion mechanisms and potential physiological roles of the dual
pathway degradation, JournalofBiologicalChemistry,vol.,
no.,pp.,.
[] M. Linnenbaum, M. Busker, J. R. Kraehling, and S. Behrends,
“Heme oxygenase isoforms dier in their subcellular tracking
during hypoxia and are dierentially modulated by cytochrome
P reductase, PLoS ONE,vol.,no.,ArticleIDe,.
[] S. W. Ryter, J. Alam, and A. M. K. Choi, “Heme oxygenase-
/carbon monoxide: from basic science to therapeutic applica-
tions, Physiological Reviews,vol.,no.,pp.,.
[] C. L. Hartseld, J. Alam, and A. M. K. Choi, “Dierential
signaling pathways of HO- gene expression in pulmonary and
systemic vascular cells, American Journal of Physiolog y,vol.,
no. , pp. L–L, .
[] J. Alam, S. Shibahara, and A. Smith, “Transcriptional activation
of the heme oxygenase gene by heme and cadmium in mouse
hepatoma cells, Journal of Biological Chemistry,vol.,no.,
pp. –, .
[] L. A. Applegate, A. Noel, G. Vile, E. Frenk, and R. M.
Tyrrell, “Two genes contribute to dierent extents to the
heme oxygenase enzyme activity measured in cultured human
skin broblasts and keratinocytes: implications for protection
against oxidant stress, Photochemistr y and Photobiology,vol.,
no.,pp.,.
[] M. Rizzardini, M. Carelli, M. R. Cabello Porras, and L. Cantoni,
“Mechanisms of endotoxin-induced haem oxygenase mRNA
accumulation in mouse liver: synergism by glutathione deple-
tion and protection by N-acetylcysteine, Biochemical Journal,
vol. , no. , pp. –, .
[] K. M. Stuhlmeier, Activation and regulation of Hsp and
Hsp,” European Journal of Biochemistry,vol.,no.,pp.
–, .
[] K. S. Callahan, “Eect of tumor necrosis factor-a and
interleukin-la on heme oxygenase- expression in human
endothelial cells, American Journal of Physiology,vol.,no.
, part , pp. H–H, .
[] R. Foresti, J. E. Clark, C. J. Green, and R. Motterlini, “iol com-
pounds interact with nitric oxide in regulating heme oxygenase-
induction in endothelial cells: involvement of superoxide and
peroxynitrite anions, Journal of Biological Chemistr y,vol.,
no. , pp. –, .
[] Y.J.Surh,J.K.Kundu,andH.K.Na,“Nrfasamasterredox
switch in turning on the cellular signaling involved in the
induction of cytoprotective genes by some chemopreventive
phytochemicals, Planta Medica,vol.,no.,pp.,
.
[] T. S. Lee and L. Y. Chau, “Heme oxygenase- mediates the anti-
inammatory eect of interleukin- in mice, Nature Medicine,
vol. , no. , pp. –, .
[] J. M. Kyriakis and J. Avruch, “Mammalian MAPK signal
transduction pathways activated by stress and inammation: a
-year update, Physiological Reviews,vol.,no.,pp.
, .
 Oxidative Medicine and Cellular Longevity
[] N. Hill-Kapturczak, C. Voakes, J. Garcia, G. Visner, H. S. Nick,
and A. Agarwal, A cis-acting region regulates oxidized lipid-
mediated induction of the human heme oxygenase- gene in
endothelial cells, Arteriosclerosis, rombosis, and Vascular
Biology,vol.,no.,pp.,.
[] E. M. Sikorski, T. Hock, N. Hill-Kapturczak, and A. Agarwal,
“e story so far: molecular regulation of the heme oxygenase-
gene in renal injury, American Journal of Physiology,vol.,
no. , pp. F–F, .
[] A. M. K. Choi and J. Alam, “Heme oxygenase-: function,
regulation, and implication of a novel stress-inducibleprotein in
oxidant-induced lung injury, American Journal of Respirator y
Cell and Molecular Biology,vol.,no.,pp.,.
[] S. J. Chapple, R. C. Siow, and G. E. Mann, “Crosstalk between
Nrf and the proteasome: therapeutic potential of Nrf inducers
in vascular disease and aging,” International Journal of Biochem-
istry & Cell Biology,vol.,no.,pp.,.
[] K. Itoh, J. Mimura, and M. Yamamoto, “Discovery of the nega-
tive regulator of Nrf, keap: a historical overview, Antioxidants
and Redox Signaling, vol. , no. , pp. –, .
[ ] R . Yu , C . Ch en , Y. Y. M o e t al . , Act i v at i on o f m i to g e n- ac t iv at e d
protein kinase pathways induces antioxidant response element-
mediated gene expression via a Nrf-dependent mechanism,
JournalofBiologicalChemistry,vol.,no.,pp.–,
.
[]J.Alam,C.Wicks,D.Stewartetal.,“Mechanismofheme
oxygenase- gene activation by cadmium in MCF- mammary
epithelial cells: role of p kinase and Nrf transcription factor,”
JournalofBiologicalChemistry,vol.,no.,pp.
, .
[] S. F. Yet, M. A. Perrella, M. D. Layne et al., “Hypoxia induces
severe right ventricular dilatation and infarction in heine
oxygenase- null mice, Journal of Clinical Investigation,vol.,
no. , pp. R–R, .
[] S. F. Yet, M. D. Layne, X. Liu et al., Absence of heme oxygenase-
exacerbates atherosclerotic lesion formation and vascular
remodeling, e FASEB Journal,vol.,no.,pp.,
.
[] K. Ishikawa, D. Sugawara, X. P. Wang et al., “Heme oxygenase-
inhibits atherosclerotic lesion formation in LDL-receptor
knockout mice, Circulation Research,vol.,no.,pp.,
.
[]X.Liu,J.Wei,D.H.Peng,M.D.Layne,andS.F.
Yet, Absence of heme oxygenase- exacerbates myocardial
ischemia/reperfusion injury in diabetic mice, Diabetes,vol.,
no. , pp. –, .
[] B. S. Zuckerbraun, T. R. Billiar, S. L. Otterbein et al., “Carbon
monoxide protects against liver failure through nitric oxide-
induced heme oxygenase , JournalofExperimentalMedicine,
vol. , no. , pp. –, .
[]B.Ke,J.W.Kupiec-Weglinski,R.Buelowetal.,“Hemeoxy-
genasegenetransferpreventsCD/Fasligand-mediated
apoptosis and improves liver allogra survival via carbon
monoxide signaling pathway,” Human Gene erapy,vol.,no.
, pp. –, .
[] L. E. Otterbein, J. K. Kolls, L. L. Mantell, J. L. Cook, J. Alam, and
A. M. K. Choi, “Exogenous administration of heme oxygenase-
by gene transfer provides protection against hyperoxia-induced
lung injury, Journal of Clinical Investigation,vol.,no.,pp.
–, .
[]R.A.Johnson,F.J.Teran,W.Durante,K.J.Peyton,andF.
K. Johnson, “Enhanced heme oxygenase-mediated coronary
vasodilation in Dahl salt-sensitive hypertension, American
Journal of Hypertension,vol.,no.,pp.,.
[] F. Gullotta, A. di Masi, M. Coletta, and P. Ascenzi, “CO
metabolism, sensing, and signaling, Biofactors,vol.,no.,
pp. –, .
[] S. W. Ryter and L. E. Otterbein, “Carbon monoxide in biology
and medicine, BioEssays, vol. , no. , pp. –, .
[] H.O.Pae,Y.Son,N.H.Kim,H.J.Jeong,K.C.Chang,and
H. T. Chung, “Role of heme oxygenase in preserving vascular
bioactive NO, Nitric Oxide,vol.,no.,pp.,.
[] S.E.Williams,S.P.Brazier,N.Babanetal.,“Astructuralmotifin
the C-terminal tail of slo confers carbon monoxide sensitivity
to human BKCa channels, Pugers Archiv European Journal of
Physiology,vol.,no.,pp.,.
[] L. E. Otterbein, F. H. Bach, J. Alam et al., Carbon monoxide
has anti-inammatory eects involving the mitogen-activated
protein kinase pathway, Nature Medicine,vol.,no.,pp.
, .
[] A. Hoetzel, T. Dolinay, S. Vallbracht et al., Carbon monoxide
protects against ventilator-induced lung injury via PPAR-𝛾and
inhibition of Egr-, American Journal of Respirator y and Critical
Care Medicine,vol.,no.,pp.,.
[] T. Dolinay, W. Wu, N. Kaminski et al., “Mitogen-activated
protein kinases regulate susceptibility to ventilator-induced
lung injury, PLoS ONE,vol.,no.,ArticleIDe,.
[] R. Gajd´
ocsy and I. Horv´
ath, “Exhaled carbon monoxide in
airway diseases: from research ndings to clinical relevance,
Journal of Breath Research,vol.,no.,ArticleID,.
[] D. E. Baranano, H. Wolosker, B. I. Bae, R. K. Barrow, S. H.
Snyder,andC.D.Ferris,“AmammalianironATPaseinducedby
iron, Journal of Biological Chemistry,vol.,no.,pp.
, .
[] A. Yachie, Y. Niida, T. Wada et al., “Oxidative stress causes
enhanced endothelial cell injury in human heme oxygenase-
deciency, Journal of Clinical Investigation,vol.,no.,pp.
–, .
[] K. D. Poss and S. Tonegawa, “Heme oxygenase is required
for mammalian iron reutilization, Proceedings of the National
Academy of Sciences of the United States of America,vol.,no.
, pp. –, .
[] R. S. Eisenstein and H. N. Munro, Translational regulation of
ferritin synthesis by iron, Enzyme,vol.,no.,pp.,
.
[]G.F.VileandR.M.Tyrrell,“Oxidativestressresultingfrom
ultraviolet A irradiation of human skin broblasts leads to
a heme oxygenase-dependent increase in ferritin, Journal of
Biological Chemistry,vol.,no.,pp.,.
[] P. O. Berberat, M. Katori, E. Kaczmarek et al., “Heavy chain
ferritin acts as an antiapoptotic gene that protects livers from
ischemia reperfusion injury, e FASEB Journal,vol.,no.,
pp.,.
[] G. F. Vile, S. Basu-Modak, C. Waltner, and R. M. Tyrrell, “Heme
oxygenase mediates an adaptive response to oxidative stress in
humanskinbroblasts,Proceedings of the National Academy of
Sciences of the United States of America,vol.,no.,pp.
, .
[] J. Kapitulnik and M. D. Maines, “e role of bile pigments in
health and disease: eects on cell signaling, cytotoxicity, and
cytoprotection, Frontiers in Pharmacology,vol.,p.,.
[] L. Vitek, “e role of bilirubin in diabetes, metabolic syndrome,
and cardiovascular diseases, Frontiers in Pharmacology,vol.,
p. , .
Oxidative Medicine and Cellular Longevity 
[] R. Stocker, Antioxidant activities of bile pigments, Antioxi-
dants and Redox Signaling,vol.,no.,pp.,.
[] T. Jansen and A. Daiber, “Direct antioxidant properties of
bilirubin and biliverdin. Is there a role for biliverdin reductase?”
Frontiers in Pharmacology,vol.,p.,.
[] B. Wegiel and L. E. Otterbein, “Go green: the anti-inammatory
eects of biliverdin reduct ase, Frontiers in Pharmacology,vol.,
p. , .
... matrix (RO×PE) = 256×128; TR=250ms; echo time (TE) = 5ms; flip angle (FA) = 20. T2* array scanning was performed with an arranged TE (6,11,16,31,36, 41ms) applying the following parameters: FOV = 35×35mm; slice thickness=1mm; matrix (RO × PE) = 128×128; TR = 250ms; FA = 20°. ...
... Thus, Leclerc et al. 5 found that CD163 knockout mice were initially protected against ICHinduced injury, but long-term injury was enhanced. Experimental studies have shown that the haptoglobin (Hp)-hemoglobin (Hb)-CD163-HO-1 pathway in macrophages is an efficient way to circumvent the Hb-induced toxicity 6 . Following physiological or pathological hemolysis, free Hb binds to Hp followed by CD163-mediated uptake of the Hb-Hp complex into microglia/macrophages. ...
Article
Background and Purpose: Early erythrolysis in the hematoma contributes to brain injury after intracerebral hemorrhage (ICH). This study investigated the effects of N-acetylheparin (NAH), a complement inhibitor, and aurin tricarboxylic acid (ATA), a membrane attack complex inhibitor, on early erythrolysis, brain iron deposition and brain injury in aged rats.
... CD163 is expressed on the surface of mature macrophages and monocytes, and functions in mediating inflammation and the removal of hemoglobin-haptoglobin complexes from the blood. 7,8 SRCR domain 5 of CD163 is an interaction site for PRRSV 9 and is encoded entirely within exon 7. Previous research demonstrated that a dual guide RNA (gRNA) approach, wherein two Cas9-gRNA complexes were targeted to the introns flanking exon 7, could result in deletion of SRCR domain 5 in pigs. 10 This editing strategy resulted in animals with PRRSV-resistant pulmonary alveolar macrophages (PAMs) and peripheral blood monocytes, and these pigs showed no signs of infection or viral replication in lung and lymph node tissue when challenged with PRRSV. ...
Article
Full-text available
Disease resistance genes in livestock provide health benefits to animals and opportunities for farmers to meet the growing demand for affordable, high-quality protein. Previously, researchers used gene editing to modify the porcine CD163 gene and demonstrated resistance to a harmful virus that causes porcine reproductive and respiratory syndrome (PRRS). To maximize potential benefits, this disease resistance trait needs to be present in commercially relevant breeding populations for multiplication and distribution of pigs. Toward this goal, a first-of-its-kind, scaled gene editing program was established to introduce a single modified CD163 allele into four genetically diverse, elite porcine lines. This effort produced healthy pigs that resisted PRRS virus infection as determined by macrophage and animal challenges. This founder population will be used for additional disease and trait testing, multiplication, and commercial distribution upon regulatory approval. Applying CRISPR-Cas to eliminate a viral disease represents a major step toward improving animal health.
... Upregulation of CD163, a hemoglobin scavenger receptor, is seen on macrophages in the direct perihematomal area and on neurons slightly more distal within the ipsilateral basal ganglia after ICH (Liu et al., 2017). Free hemoglobin binds to haptoglobin and is taken up into cells through CD163 mediated transport (Thomsen et al., 2013). Within the cell, hemoglobin is degraded into heme which is then degraded into carbon monoxide, biliverdin and Fe 2+ by heme-oxygenase (HO)-1 or 2 (Siow et al., 1999). ...
Article
Intracerebral hemorrhage (ICH) is a significant cause of death and disability and current treatment is limited to supportive measures to reduce brain edema and secondary hematoma expansion. Current evidence suggests that the complement cascade is activated early after hemorrhage and contributes to brain edema/injury in multiple ways. The aim of this review is to summarize the most recent literature about the role of the complement cascade after ICH. Primary literature demonstrating complement mediated brain edema and neurologic injury through the membrane attack complex (MAC) as well as C3a and C5a are reviewed. Further, attenuation of brain edema and improved functional outcomes are demonstrated after inhibition of specific components of the complement cascade. Conversely, complement also plays a significant role in neurologic recovery after ICH and in other neurologic disorders. We conclude that the role of complement after ICH is complex. Understanding the role of complement after ICH is essential and may elucidate possible interventions to reduce brain edema and injury.
... 3 Ferritin is induced to bind intracellular iron. 19 Immunohistochemistry demonstrated that the HO-1positive cells were significantly fewer in the hematoma, perihematomal tissue, and ipsilateral BG tissue in C3-deficient mice on day 3 after ICH (hematoma: 251±102 vs. 607±18 cells/mm 2 in C3-sufficient mice, P < 0.01; perihematomal tissue: 637±143 vs. 1025±182 cells/mm 2 in C3-sufficient mice, P < 0.01; ipsilateral BG: 376±125 vs. 557±82 in C3-sufficient mice, P < 0.01; Figure 3A). ...
Article
Background and Purpose: Early erythrolysis occurs within the hematoma following intracerebral hemorrhage (ICH) and the release of erythrocyte cytoplasmic proteins such as hemoglobin and peroxiredoxin 2 (Prx2) can cause brain injury. Complement activation can induce erythrolysis. This study determined the function of complement component 3 (C3) in erythrolysis in hematoma and brain injury after ICH in mice.
... Due to their hydrophobic nature, heme and hemin are incorporated into biological membranes and cause lipid peroxidation and cell death. To prevent this harmful scenario, macrophages clear the hemoglobin-haptoglobin complex via their CD163 receptor and hemin with CD36 receptor and upregulate the expression of HMOX1, which degrades the porphyrin ring 42,43 . The heme degradation products biliverdin and bilirubin have antioxidant and anti-inflammatory effects, contributing to tissue repair after injury 44 . ...
Article
Full-text available
Extensive mechanical stress frequently causes micro-traumas in skeletal muscle, followed by a regeneration period. The effective removal of dead myofibers is a prerequisite for proper regeneration, and several cell types, including professional phagocytes, were reported to be active in this process. Myoblasts express several molecules of the phagocytic machinery, such as BAI1, stabilin-2, and TAM (Tyro3, Axl, Mertk) tyrosine kinase receptors, but these molecules were reported to serve primarily cell fusion and survival, and their role in the phagocytosis was not investigated. Therefore, we aimed to investigate the in vitro phagocytic capacity of the C2C12 mouse myoblast cell line. RNA sequencing data were analyzed to determine the level and changes of phagocytosis-related gene expression during the differentiation process of C2C12 cells. To study the phagocytic capacity of myoblasts and the effect of dexamethasone, all-trans retinoic acid, hemin, and TAM kinase inhibitor treatments on phagocytosis, C2C12 cells were fed dead thymocytes, and their phagocytic capacity was determined by flow cytometry. The effect of dexamethasone and all-trans retinoic acid on phagocytosis-related gene expression was determined by quantitative PCR. Both undifferentiated and differentiated cells engulfed dead cells being the undifferentiated cells more effective. In line with this, we observed that the expression of several phagocytosis-related genes was downregulated during the differentiation process. The phagocytosis could be increased by dexamethasone and all-trans retinoic acid and decreased by hemin and TAM kinase inhibitor treatments. Our results indicate that myoblasts not only express phagocytic machinery genes but are capable of efficient dead cell clearance as well, and this is regulated similarly, as reported in professional phagocytes.
... Haptoglobin (Hp) functions as an effective Hb scavenger by binding irreversibly to Hb and preventing haem release. Hp is present in blood plasma within the range of 0.3-3 mg/mL Kazmi et al., 2019;Thomsen et al., 2013). However, the large multimeric Hp proteins do not easily cross the blood-brain barrier, hence the concentration of Hp is much lower in the central nervous system compared to the systemic circulation , and Hp is rapidly depleted in the cerebrospinal fluid (CSF) after SAH (Akeret et al., 2021). ...
Article
Full-text available
During subarachnoid haemorrhage, a blood clot forms in the subarachnoid space releasing extracellular haemoglobin (Hb), which causes oxidative damage and cell death in surrounding tissues. High rates of disability and cognitive decline in SAH survivors are attributed to loss of neurons and functional connections during secondary brain injury. Haptoglobin sequesters Hb for clearance, but this scavenging system is overwhelmed after a haemorrhage. Whilst exogenous haptoglobin application can attenuate cytotoxicity of Hb in vitro and in vivo, the functional effects of sub‐lethal Hb concentrations on surviving neurons and whether cellular function can be protected with haptoglobin treatment remain unclear. Here we use cultured neurons to investigate neuronal health and function across a range of Hb concentrations to establish the thresholds for cellular damage and investigate synaptic function. Hb impairs ATP concentrations and cytoskeletal structure. At clinically relevant but sub‐lethal Hb concentrations, we find that synaptic AMPAR‐driven currents are reduced, accompanied by a reduction in GluA1 subunit expression. Haptoglobin co‐application can prevent these deficits by scavenging free Hb to reduce it to sub‐threshold concentrations and does not need to be present at stoichiometric amounts to achieve efficacy. Haptoglobin itself does not impair measures of neuronal health and function at any concentration tested. Our data highlight a role for Hb in modifying synaptic function in surviving neurons, which may link to impaired cognition or plasticity after SAH and support the development of haptoglobin as a therapy for subarachnoid haemorrhage. image
... CD163 is known as the HP receptor. The interaction between HP and CD163 increases the expression of heme oxygenase-1 (HO-1), which reduces inflammation and oxidative stress [19]. CD163 has also been shown to differentiate macrophage type 2 (M2) which, in turn, decreases inflammation in COPD. ...
Article
Full-text available
Background Chronic lung diseases are characterized by impaired lung function. Given that many diseases have shared clinical symptoms and pathogenesis, identifying shared pathogenesis can help the design of preventive and therapeutic strategies. This study aimed to evaluate the proteins and pathways of chronic obstructive pulmonary disease (COPD), asthma, idiopathic pulmonary fibrosis (IPF), and mustard lung disease (MLD). Methods and results After collecting the data and determining the gene list of each disease, gene expression changes were examined in comparison to healthy individuals. Protein–protein interaction (PPI) and pathway enrichment analysis were used to evaluate genes and shared pathways of the four diseases. There were 22 shared genes, including ACTB, AHSG, ALB, APO, A1, APO C3, FTH1, GAPDH, GC, GSTP1, HP, HSPB1, IGKC, KRT10, KRT9, LCN1, PSMA2, RBP4, 100A8, S100A9, TF, and UBE2N. The major biological pathways in which these genes are involved are inflammatory pathways. Some of these genes activate different pathways in each disease, leading to the induction or inhibition of inflammation. Conclusion Identification of the genes and shared pathways of diseases can contribute to identifying pathogenesis pathways and designing preventive and therapeutic strategies.
Article
Macrophages in the endometrium promote receptivity and implantation by secreting proinflammatory cytokines and other factors like fractalkine (FKN). Macrophages are closely linked to regulating iron homeostasis and can modulate iron availability in the tissue microenvironment. It has been revealed that the iron metabolism of the mother is crucial in fertility. Iron metabolism is strictly controlled by hepcidin, the principal iron regulatory protein. The inflammatory cytokines can modulate hepcidin synthesis and, therefore, the iron metabolism of the endometrium. It was proven recently that FKN, a unique chemokine, is implicated in maternal–fetal communication and may contribute to endometrial receptivity and implantation. In the present study, we investigated the effect of activated THP‐1 macrophages and FKN on the iron metabolism of the HEC‐1A endometrial cells. We established a noncontact coculture with or without recombinant human FKN supplementation to study the impact of the macrophage‐derived factors and FKN on the regulation of hepcidin synthesis and iron release and storage of endometrial cells. Based on our findings, the conditioned medium of the activated macrophages could modify hepcidin synthesis via the nuclear factor kappa‐light‐chain‐enhancer of activated B cells, the signal transducer and activator of transcription 3, and the transferrin receptor 2/bone morphogenetic protein 6/suppressor of mothers against decapentaplegic 1/5/8 signaling pathways, and FKN could alter this effect on the endometrial cells. It was also revealed that the conditioned macrophage medium and FKN modulated the iron release and storage of HEC‐1A cells. FKN signaling may be involved in the management of iron trafficking of the endometrium by the regulation of hepcidin. It can contribute to the iron supply for fetal development at the early stage of the pregnancy.
Article
Background Patients with sickle cell disease (SCD) experience intravascular hemolysis, leading to elevated plasma heme levels. This phenomenon has been associated with increased priapism in men with SCD. The heme group can be metabolized by heme oxygenase (HO), generating carbon monoxide (CO), which is known to promote smooth muscle relaxation via soluble guanylyl cyclase (sGC)–cyclic guanosine monophosphate (cGMP). However, the effects of heme on the relaxation responses of corpus cavernosum (CC) have not been investigated. Objectives To evaluate the functional and biochemical effects of the heme group on mouse CC smooth muscle in vitro. Materials and methods Male C57BL/6 mice were used. CC tissues were mounted in organ baths. Measurement of cGMP in mice CC was evaluated. Results The cumulative addition of heme concentrations promoted the relaxation of CC. HO inhibitor (1J, 100 μM) or sGC inhibitor (ODQ, 10 μM) blocked the relaxing effect of the heme group. Pre‐incubation of CC with heme (100 μM) enhanced relaxation induced by acetylcholine, sodium nitroprusside, and nitrergic relaxation (electrical field stimulation), which was abolished by 1J or ODQ. The heme group increased the cGMP production in CC, which was abolished by 1J or ODQ. cGMP levels were significantly higher in CC treated with heme, and pre‐incubation with compound 1J or ODQ abolished the effect of heme in raising cGMP levels. Discussion and conclusion The HO–CO–sGC–cGMP pathway appears to play a crucial role in promoting CC relaxation. Our study provides novel insight into the role of group heme in CC relaxation and its potential contribution to priapism in SCD. Heme may serve as a pharmacological target for new therapies to prevent priapism.
Article
Dysregulation of iron metabolism has been associated with impaired chronic wound healing. However, changes in iron metabolism have yet to be reported in pressure injuries, a type of chronic wound. In this study, we aimed to investigate changes in iron metabolism and associated regulatory mechanisms in pressure injuries. We collected tissue biopsies and data from twenty consenting stage IV‐ pressure injuries patients and five non‐pressure injuries patients hospitalized at the Affiliated Hospital of Qingdao University between March 2021 and June 2021. In addition, we measured the iron content by inductively coupled plasma mass spectrometry and Prussian blue staining in deep tissue pressure injury mouse models. An Enzyme‐linked immune sorbent assay measured the expression of ferritin, ferroportin‐1, and transferrin. Immunofluorescence staining, High‐throughput transcriptome sequencing, Western blot, and RT‐qPCR further analyzed the fundamental mechanisms regulating iron metabolism. In this study, we observed numerous inflammatory cells infiltrating the marginal tissues of stage IV pressure injury patients and in deep tissue pressure injury models. The expression levels of proinflammatory factors, such as inducible nitric oxide synthase and interleukin‐6, were significantly increased (p<0.05). The iron level was proportional to the degree of progression, with the most significant change appearing on the third day in deep tissue pressure injury models (p<0.05). Enzyme‐linked immune sorbent assay results suggested abnormal gene expression was related to iron metabolism, including a substantial increase in ferritin and a significant decrease in the expression of ferroportin‐1 (p<0.05). In addition, immunofluorescence staining and Western blot showed that the expression of macrophage membrane receptor CD163 was abnormally elevated (p<0.05). Both high‐throughput transcriptome sequencing and qRT‐PCR results suggested aberrant expression of the CD163/Homx‐1‐mediated signaling pathway. Dysfunctional iron metabolism was suggested to be related to the aberrant CD163/Homx‐1 signaling pathway in deep tissue pressure injury models. This article is protected by copyright. All rights reserved.
Article
Full-text available
Intravascular hemolysis is a hallmark event in the immunopathology of malaria that results in increased systemic concentrations of free hemoglobin (Hb). The oxidation of Hb by free radicals causes the release of heme, which amplifies inflammation. To circumvent the detrimental effects of free heme, hosts have developed several homeostatic mechanisms, including the enzyme haptoglobin (Hp), which scavenges cell-free Hb, the monocyte receptor CD163, which binds to Hb-Hp complexes, and heme oxygenase-1 (HO-1), which degrades intracellular free heme. We tested the association between these three main components of the host response to hemolysis and susceptibility to malaria in a Brazilian population. The genetic profiles of the HMOX1 and Hp genes and the plasma levels of a serum inflammatory marker, the soluble form of the CD163 receptor (sCD163), were studied in 264 subjects, including 78 individuals with symptomatic malaria, 106 individuals with asymptomatic malaria, and 80 uninfected individuals. We found that long (GT)n repeats in the microsatellite polymorphism region of the HMOX1 gene, the Hp2 allele, and the Hp2.2 genotype were associated with symptomatic malaria. Moreover, increased plasma concentrations of heme, Hp, HO-1, and sCD163 were associated with susceptibility to malaria. The validation of these results could support the development of targeted therapies and aid in reducing the severity of malaria.
Article
Full-text available
Formation of the haptoglobin (Hp)-hemoglobin (Hb) complex in human plasma leads to a high affinity recognition by the endocytic macrophage receptor CD163. A fast segregation of Hp-Hb from CD163 occurs at endosomal conditions (pH <6.5). The ligand binding site of CD163 has previously been shown to involve the scavenger receptor cysteine-rich (SRCR) domain 3. This domain and the adjacent SRCR domain 2 of CD163 contain a consensus motif for a calcium-coordinated acidic amino acid triad cluster as originally identified in the SRCR domain of the scavenger receptor MARCO. Here we show that site-directed mutagenesis in each of these acidic triads of SRCR domains 2 and 3 abrogates the high affinity binding of recombinant CD163 to Hp-Hb. In the ligand, Hp Arg-252 and Lys-262, both present in a previously identified CD163 binding loop of Hp, were revealed as essential residues for the high affinity receptor binding. These findings are in accordance with pairing of the calcium-coordinated acidic clusters in SRCR domains 2 and 3 with the two basic Arg/Lys residues in the Hp loop. Such a two-point electrostatic pairing is mechanistically similar to the pH-sensitive pairings disclosed in crystal structures of ligands in complex with tandem LDL receptor repeats or tandem CUB domains in other endocytic receptors. Background: CD163 mediates endocytosis of haptoglobin-hemoglobin complexes formed upon intravascular hemolysis. Results: Acidic CD163 residues and basic haptoglobin residues mediate receptor-ligand binding in humans. Conclusion: A two-point electrostatic pairing between Ca²⁺-coordinated acidic clusters in receptors and basic ligand residues explains high-affinity CD163-(haptoglobin-hemoglobin) binding. Significance: The data add further evidence that Ca²⁺-dependent two-point electrostatic pairing is a common mechanism for coupling ligands to endocytic receptors.
Article
Heme iron exacerbates oxidant damage by catalyzing the production of free radicals. Heme oxygenase is the rate-limiting enzyme involved in heme catabolism. An inducible form of heme oxygenase, heme oxygenase-1 (HO-1), is upregulated in oxidant and inflammatory settings, and recent work suggests that HO-1 induction may serve a protective function against oxidant injury. The ability of the endogenous inflammatory mediators, interleukin (IL)-la, tumor necrosis factor-a (TNF-a), and IL-6, to enhance HO-1 expression in cultured human endothelial cells was examined in this study. HO-1 mRNA and protein expression were upregulated by IL-la and TNF-a exposure but not by IL-6. Induction of HO-1 mRNA by IL-la and TNF-a occurred in a concentration- and time-dependent fashion, with maximal expression occurring by 4 h for both cytokines. Induction depended on protein synthesis and occurred at the transcriptional level. Inhibition of the AP-1 transcription factor with curcumin decreased the cytokine induction of HO-1 mRNA, suggesting the involvement of this transcription factor in cytokine signaling of HO-1. The results of this study indicate that the endogenous inflammatory cytokines IL-la and TNF-a induce HO-1 in endothelial cells, providing further evidence that HO-1 may be an important cellular response to inflammatory stress. cytokine; inflammation; heme oxygenase.
Article
Haptoglobin (Hp) is a plasma glycoprotein, the main biological function of which is to bind free hemoglobin (Hb) and prevent the loss of iron and subsequent kidney damage following intravascular hemolysis. Haptoglobin is also a positive acute-phase protein with immunomodulatory properties. In humans, the HP locus is polymorphic, with two codominant alleles (HP1 and HP2) that yield three distinct genotypes/phenotypes (Hp1-1, Hp2-1 and Hp2-2). The corresponding proteins have structural and functional differences that may influence the susceptibility and/or outcome in several diseases. This article summarizes the available data on the structure and functions of Hp and the possible effects of Hp polymorphism in a number of important human disorders.
Article
Background: Cardiovascular tissues express heme oxygenase (HO), which metabolizes heme to form carbon monoxide (CO). Carbon monoxide promotes relaxation of coronary vascular smooth muscle. Increased HO-1 expression provides cardioprotection during certain pathologic conditions. On a high salt (HS) diet Dahl salt-sensitive (DS) rats develop hypertension that is accompanied by left ventricular hypertrophy, whereas Dahl salt-resistant rats (DR) do not. This study tests the hypothesis that cardiac HO-1 expression is increased in DS rats with salt-induced hypertension and provides cardioprotection by promoting coronary vasodilation.
Article
The haptoglobin phenotypes of Sudanese patients with complicated and uncomplicated falciparum malaria, and those of uninfected randomly selected individuals, were determined by electrophoresis of sera on polyacrylamide gels followed by benzidine staining of the gels. Among 273 malaria patients, the proportions with haptoglobin phenotypes (1-1), (2-1) and (2-2) were 60·8%, 29·7% and 9·5%, respectively, and in 72 cerebral malaria patients the proportions were 63·9%, 29·2% and 6·9%. The distribution among 208 control individuals was 26·0%, 55·8% and 18·3%, respectively. The difference between patients and controls was highly significant (P < 0·001). The distribution of the different haptoglobin phenotypes among the randomly selected group of 208 Sudanese individuals was comparable to that in many other populations. The results suggest that the haptoglobin phenotype (1-1) is associated with susceptibility to falciparum malaria and the development of severe complications; alternatively, the other phenotypes may confer resistance.
Article
70 in lung tissue. In contrast, CO application prevented lung injury during ventilation, inhibited stress-gene up-regulation, and de- creased lung neutrophil infiltration. These effects were preceded by the inhibition of ventilation-induced cytokine and chemokine production. Furthermore, CO prevented the early ventilation- dependentup-regulationofearly growthresponse-1 (Egr-1).Egr-1- injury Mechanical ventilation, routinely required for the clinical treat- ment of respiratory failure, can cause ventilator-induced lung injury (VILI), and aggravate the progression of acute respira- tory distress syndrome. Despite the implementation of pro- tective ventilation strategies (1-3), involving the reduction of tidal volume, the increase of positive end-expiratory pressure, or lung recruitment maneuvers, mechanical ventilation con- tributes to an unacceptably high mortality in these patients. Therefore, additional strategies must be developed to further decrease the risk of ventilator-associated lung injury. The development of VILI involves a complex orchestration of biotraumatic events. Ventilation, which imposes mechanical
Article
Anemia is a frequently encountered problem in the critically ill patient. The inability to compensate for anemia includes several mechanisms, collectively referred to as anemia of inflammation: reduced production of erythropoietin, impaired bone marrow response to erythropoietin, reduced iron availability, and increased red blood cell (RBC) clearance. This review focuses on mechanisms of RBC clearance during inflammation. We state that phosphatidylserine (PS) expression in inflammation is mainly enhanced due to an increase in ceramide, caused by an increase in sphingomyelinase activity due to either platelet activating factor, tumor necrosis factor-α, or direct production by bacteria. Phagocytosis of RBCs during inflammation is mediated via RBC membrane protein band 3. Reduced deformability of RBCs seems an important feature in inflammation, also mediated by band 3 as well as by nitric oxide, reactive oxygen species, and sialic acid residues. Also, adherence of RBCs to the endothelium is increased during inflammation, most likely due to increased expression of endothelial adhesion molecules as well as PS on the RBC membrane, in combination with decreased capillary blood flow. Thereby, clearance of RBCs during inflammation shows similarities to clearance of senescent RBCs, but also has distinct entities, including increased adhesion to the endothelium.