You are on page 1of 10

Am J Physiol Lung Cell Mol Physiol 324: L169–L178, 2023.

First published January 3, 2023; doi:10.1152/ajplung.00127.2022

PERSPECTIVES

The ultimate tradeoff: how red cell adaptations to malaria alter the host
response during critical illness
Jane Dobkin,1 Ling Wu,2,3 and Nilam S. Mangalmurti2,3
1
Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; 2Division of Pulmonary, Allergy and
Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and 3Lung Biology
Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania

Abstract
The human immune system evolved in response to pathogens. Among these pathogens, malaria has proven to be one of the
deadliest and has exerted the most potent selective pressures on its target cell, the red blood cell. Red blood cells have
recently gained recognition for their immunomodulatory properties, yet how red cell adaptations contribute to the host response
during critical illness remains understudied. This review will discuss how adaptations that may have been advantageous for host
survival might influence immune responses in modern critical illness. We will highlight the current evidence for divergent host re-
silience arising from the adaptations to malaria and summarize how understanding evolutionary red cell adaptations to malaria
may provide insight into the heterogeneity of the host response to critical illness, perhaps driving future precision medicine
approaches to syndromes affecting the critically ill such as sepsis and acute respiratory distress syndrome (ARDS).

ARDS; COVID-19; malaria; red blood cell; sepsis

INTRODUCTION in erythrocytes. Infected erythrocytes then burst, allowing


the parasite to invade other neighboring erythrocytes (6).
Malaria has threatened human existence for millennia. It is It comes as no surprise that the host target of malarial
uniquely positioned to be a driver of natural selection, given selection pressures is the same cell at which parasite
its propensity to infect children and cause fatality before sequestration and replication occurs—the red blood cell.
reproductive age. In part, survival depends on red blood cell Decades of work have been dedicated to investigating the
(RBC) mutations that confer innate resistance to the parasite link between malaria and known alterations in gas-
(1–3). J.B.S. Haldane first noted in the 1940s that the geo- exchanging properties of the RBC. Emerging evidence sug-
graphical distribution of sickle cell disease, the most well- gests further adaptations to virtually every component of
studied red blood cell disorder of the time, aligned with the the RBC, from membrane proteins to oxidative and glyco-
tropical regions where malaria was endemic. He posited lytic enzymes to the newly discovered expression of
his since-termed “Malaria Hypothesis” that this overlap immune receptors on RBCs (Fig. 1; Table 1).
was no coincidence, but rather selection in those regions Critical illnesses such as sepsis and acute respiratory dis-
for heterozygotes who are more resistant to infection with tress syndrome (ARDS) portend high morbidity and mortal-
the Plasmodium species that cause malaria (4). A.C. ity with a multitude of mechanisms that collectively lead to
Allison lent credence to this hypothesis less than a decade profound inflammation, tissue injury, and ultimately organ
later in a series of experiments that proved that the sickle failure. More and more, as we unveil these distinctive mech-
cell polymorphism was balanced such that “the heterozy- anisms, we start to recognize the diverse outcomes that stem
gote has an advantage over either homozygote” (5). Since from patient heterogeneity. For years, RBC dysfunction was
that time, malaria has become one of the most extensively perceived to accompany critical illness as a byproduct of ex-
studied and most elegant examples of an infectious force cessive inflammation that resulted in RBC membrane distor-
as the agent of natural selection. tion, oxidative disequilibrium, and insufficient oxygen
Malarial infection originates when female anopheles delivery (38). However, recent studies have demonstrated
mosquitoes bite and inject plasmodium parasites (sporo- that RBCs play an active role in modulating diverse immune
zoites) into the bloodstream. These sporozoites travel first functions critical for host defense, including chemokine reg-
to the liver where they undergo asexual multiplication, ulation and nucleic acid scavenging. This long-overlooked
before emerging as merozoites, which invade and multiply erythrocyte contribution to host defense implores further

J. Dobkin and L. Wu contributed equally to this work.


Correspondence: N. S. Mangalmurti (nspatel@pennmedicine.upenn.edu).
Submitted 18 April 2022 / Revised 9 December 2022 / Accepted 22 December 2022

http://www.ajplung.org 1040-0605/23 Copyright © 2023 the American Physiological Society. L169


Downloaded from journals.physiology.org/journal/ajplung (189.203.131.199) on March 2, 2024.
RED CELL ADAPTATIONS AND CRITICAL ILLNESS

Figure 1. Schematic overview of known and potential red blood cell adaptations to malaria. Known malaria adaptions include Duffy antigen receptor for
chemokines (DARC), which modulates chemokine availability and neutrophil recruitment; hemoglobinopathies (sickle cell trait/disease and thalassemia);
and enzymopathies (G6PD and pyruvate kinase deficiency), which can lead to hemolysis and subsequent free heme release, promoting inflammation
and increase susceptibility to infection. Probable malarial adaptations include: ABO glycoproteins, which determine major ABO blood group, Toll-like re-
ceptor 9 (TLR9), which binds nucleic acids and mediates clearance of RBCs during inflammation, and miRNAs, which are abundant in RBCs but the func-
tions of many remain undefined. G6PD, glucose-6-phosphate dehydrogenase; RBCs, red blood cells. [Image created with BioRender.com and
published with permission.]

understanding of its role in critical illness, namely, not as KNOWN MALARIAL ADAPTATIONS AND
mere vessels for gas exchange but as active participants in THEIR RELEVANCE TO CRITICAL ILLNESS
immunoregulation. The coronavirus disease 2019 (COVID-
19) pandemic has highlighted the remarkable heterogeneity The Erythrocyte Chemokine Receptor ACKR1—Duffy
of the host response to a specific pathogen. Given the impor- Antigen Receptor for Chemokines
tance of the host response to infection in mediating out-
comes and treatment response, it is imperative that a Perhaps best known as the receptor by which Plasmodium
comprehensive understanding of all potential modulators of vivax species gain entry into red blood cells, the Duffy
the innate immune response is obtained. Here, we will Antigen Receptor for Chemokines (DARC) was initially
review the emerging evidence for alterations in RBC immune reported as a chemokine binding protein on erythrocytes.
function as a byproduct of the evolutionary response to Though unnamed and unidentified at that time, it was first
malaria and will highlight some of the major advances over described in a 1950 case report of a hemophilic patient
the past century in understanding RBC immunoregulatory who suffered a hemolytic transfusion reaction due to an
biology. Equipped with this knowledge, we will be able to antibody to an unknown substance on red blood cells
better understand how RBC immune function contributes to (RBCs) (39). Subsequently, RBCs were demonstrated to
heterogeneous host responses in the face of pathogenic or carry a surface protein that binds the chemokine interleu-
sterile insults that underlie critical illnesses such as sepsis kin- 8 (IL-8), diminishing the concentration of soluble IL-8
and ARDS. in blood plasma samples and thereby dampening immune

L170 AJP-Lung Cell Mol Physiol  doi:10.1152/ajplung.00127.2022  www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (189.203.131.199) on March 2, 2024.
RED CELL ADAPTATIONS AND CRITICAL ILLNESS

Table 1. Non-gas exchanging functions of RBCs and hypothesized role in critical illness
Hypothesized or
Proven
Adaptation RBC Feature Function Adaptation to Malaria Consequence to Critical Illness
Proven Heme Generates reactive oxygen Hemoglobinopathies/thalasse- Increased susceptibility to nonma-
species: activates TLR4 mia (8) larial infection (9, 10), anemia/
Amplifies cytokine produc- transfusion dependence (11)
tion in the presence of
LPS (7)
Duffy Antigen Receptor Binds chemokines: maintains Downregulation: decreased par- Increased susceptibility to ARDS
for Chemokines chemokine gradient (12) asite entry (13, 14) (15, 16)
Binds pathogens: entry point
for P. vivax (13)
Glucose-6-Phosphotase Involved in pathway to con- Deficiency: increased intracellu- COVID-19 severity (19)
sume reactive oxygen spe- lar oxidative stress decreases
cies (17) parasite survival (18)
Pyruvate kinase Catalyzes rate-limiting step in Deficiency: decreased invasion Hemolytic anemia/transfusion de-
glycolysis to produce intra- by P. falciparum and pendency, functional asplenia
cellular energy in nonmito- increased phagocytosis of (20)
chondrial cells (mature ring- and mature-stage-
RBCs) (20) infected RBCs (20, 21)
Hypothesized Glycophorins Binds pathogens: entry point Dantu variant: structural variant Potential predisposition to lysis/
for P. falciparum (22) in chromosome 4 region, thrombosis
which increases RBC surface
tension and decreases mero-
zoite invasion (23)
Complement Receptor 1 Binds P. falciparum-induced Deficiency: increased cerebral Increased RBC-bound complement
ligand (24, 25) malaria (26) fragments in COVID-19 and non-
Proinflammatory regulator Deficiency: reduced rosetting COVID-19 sepsis (27)
abundant on RBCs (27) conferring protection (25)
Toll-like Receptor 9 Binds nucleic acids (28, 29) Deficiency/gene polymorphisms Decreased capacity to bind mtDNA
Involved in P. yoeli infection to ameliorate lung injury (28)
(30, 31)
ABO Glycoproteins Surface proteins on RBCs re- Blood group O demonstrates dif- Blood group A may confer
sponsible for determining ferent propensity to bind to increased risk for ARDS in crit-
ABO blood group (32) Duffy binding-like regions of ically ill patients and associated
infected erythrocytes (33–36) with increased risk of AKI (37)
AKI, acute kidney injury; ARDS, acute respiratory distress syndrome; COVID-19, coronavirus disease 2019; LPS, lipopolysaccharide;
RBCs, red blood cells.

signaling (12). In 1993, Horuk et al. (13) united these two in- on erythrocytes, thereby diminishing its chemokine-binding
dependent findings and showed that the Duffy antigen re- capacity (15). As such, these DARC-low erythrocytes are less
ceptor, used by Plasmodium vivax for erythrocyte cell capable of dampening local tissue inflammation following
entry, was the same RBC receptor that bound IL-8. Since LPS-induced lung injury.
that time, DARC has been found to bind multiple inflam- Consistent with the theory of malaria as a potent driver of
matory chemokines with high affinity (40, 41). human evolution, the DARC-null allele is present in as high
DARC belongs to a family of chemokine receptors that as 90%–95% of sub-Saharan Africans in regions with the
lack a G-coupled protein to mediate intracellular signaling, highest threat of P. vivax (14). In the United States, up to
rendering the chemokine inaccessible to circulating immune 68% of African Americans have the DARC-null allele (14).
cells (40–42). As such, DARC plays a significant immunore- One observational study showed that patients with the
gulatory role by sequestering chemokines from immune Duffy-null phenotype with acute lung injury were at
cells such as neutrophils and acts as a reservoir for future increased risk of mortality at 60 days, had increased number
release (43–46). In preclinical studies using lipopolysac- of days on mechanical ventilation, and had increased organ
charide (LPS)-induced lung injury, erythrocyte DARC was failure compared with the Duffy-positive counterparts (16).
found to dampen the local chemokine effects by soaking Of note, Duffy-positive African Americans had similar clini-
up chemokines in the lung microvasculature for release in cal outcomes compared with those of European descent.
the systemic circulation, preventing polymorphonuclear Though larger studies have yet to validate these findings,
leukocyte infiltration into the alveolar space to cause lung and plasma cytokines have yet to confirm the mechanism,
injury (45). these observations complement preclinical data denoting
It is thus of no surprise that RBC-derived cytokines play a erythrocyte DARC as an essential player that tempers local
role in transfusion-related lung injury. Lysis of transfused tissue inflammation. Thus, the same adaptation that pro-
RBCs that contain chemokines can lead to profound lung vides survival benefits to the threat of malaria infection may
inflammation and result in antibody negative-transfusion- confer a higher risk of mortality when facing critical illness
related acute lung injury (TRALI) (47). Interestingly, pro- in an ICU setting, merely by altering the RBC’s ability to
longed RBC storage can result in decreased DARC expression sequester and transport inflammatory chemokines.

AJP-Lung Cell Mol Physiol  doi:10.1152/ajplung.00127.2022  www.ajplung.org L171


Downloaded from journals.physiology.org/journal/ajplung (189.203.131.199) on March 2, 2024.
RED CELL ADAPTATIONS AND CRITICAL ILLNESS

Hemoglobinopathies shown to coincide with malaria-endemic regions, leading to


hypotheses that these mutations also confer selective advant-
Hemoglobin has evolved to adapt to malarial pressures, age against deadly malaria infections (55).
rendering erythrocytes less hospitable to these invading par- One proposed mechanism for that protection against
asites. However, under acute stress, hemoglobinopathies malaria is increased susceptibility to P. vivax and P. falcipa-
lead to hemolysis and subsequent release of chemokines and rum during the neonatal period, likely due to increased
free heme into the circulation, promoting inflammation. erythrocytosis, acting as a natural vaccine and providing
more enhanced protection in later life (56). Others have
HbSS. shown that parasitized a-thalassemic erythrocytes express
Of the hemoglobin adaptations to malaria-inducing patho- higher levels of novel surface antigens (which are currently
gens, Hemoglobin SS (HbSS) is the most well-studied. HbSS ill-defined) than wild-type parasitized erythrocytes, with a
arises from a single point mutation in the b-globin chain of corresponding increase in antiparasitic antibody binding in
the hemoglobin molecule. The resultant amino acid substi- vitro (57, 58). This enhanced detection and subsequent
tution of this mutation leads to polymerization of globin enhanced clearance of parasitized RBCs may explain how
chains, which causes numerous and devastating sequelae of thalassemia protects against malaria.
sickle cell disease (48). There have been countless studies Despite protection against malaria, thalassemia carriers are
confirming that the persistence of this hemoglobin mutation at increased infection risks due to several qualitative and func-
is due to the protective advantage against P. falciparum con- tional defects in the immune system rendering them suscepti-
ferred in heterozygotes of the mutation (49). ble to pathogens such as Klebsiella pneumoniae and Yersinia
The sequelae of sickle cell disease are known to impact host enterocolitica infections (9). These defects range from defec-
immunity indirectly via numerous vaso-occlusive mecha- tive T and B lymphocyte functions to altered chemotactic and
nisms and splenectomy. Free heme released from hemolysis phagocytotic processes to complement system deficiencies (9).
during vaso-occlusive crisis is extremely proinflammatory Indirect consequences of chronic anemia also led to mortality
and can lead to endothelial damage (50). Sickled erythrocytes and morbidity from increased exposure to transfusion-trans-
have also been shown to retain mitochondria, which can pro- mitted pathogens like Hepatitis C, chronic transfusion-related
vide immunologically active mitochondria DNA (mtDNA) iron overload, and therapeutic splenectomies (11). Given this
that ultimately leads to endothelial damage. Whether mito- increased infection burden and mortality, this genetic adapta-
chondria retention is an advantageous adaption to malaria or tion to malaria has resulted in a less fit host if they live in
a byproduct of abnormal erythroid development remains to regions with unlikely threat of malaria.
be seen (51, 52). Nevertheless, mtDNA is elevated in patients
with sickle cell disease and further increases during sickle cell Enzymopathies
crisis. Notably, mtDNA is also elevated in ARDS and is associ- G6PD deficiency.
ated with increased mortality (53). It has long been appreci- Another well-studied RBC adaptation to malarial infection is
ated that patients with sickle cell disease are at higher risk of glucose-6-phosphate dehydrogenase (G6PD) deficiency. G6PD
severe disease and hospitalization from bacterial and viral is an enzyme in the pathway that consumes oxygen-free radi-
infections and confer a higher risk for ICU admissions. cals within RBCs, thereby minimizing oxidative stress. This X-
Whether the level of mtDNA in patients with sickle cell dis- linked, inherited RBC disorder has increased prevalence in
ease correlates with a higher risk of acute chest syndrome or regions with the highest malaria burden, and the prevalent
predisposition to severe ARDS has yet to be determined. form of G6PD deficiency in Africa has been demonstrated to
Despite long-held assumptions that the sickle cell trait is a correlate with a 50% reduced risk of severe malaria (17). The
silent protector against malaria, emerging evidence suggests hypothesized mechanism of protection is that the increase in
that heterozygotes may have altered susceptibility to critical oxidative stress seen in G6PD-deficient RBCs makes them a
illness. Sickle cell trait is usually asymptomatic but in rare less hospitable host for malaria-causing parasites.
instances, severe dehydration or hypoxia can lead to erythro- Although this adaptation offers protection from the poten-
cyte sickling, vaso-occlusive crisis, and exertional rhabdo- tially lethal pathogen, it comes at a cost. The oxidative stress
myolysis. Notably, a recent study found increased risk of leads to early RBC breakdown and, in situations of stress or
mortality and acute kidney injury after SARS-CoV-2 infec- exposure, widespread hemolysis, leading to free heme and
tion in patients of African ancestry with sickle cell trait (54). chemokine release. Those with G6PD deficiency have also
Dedicated investigation into the impact of sickle cell trait on been shown to fare worse in the face of critical illness during
red blood cell immune function would help to better under- COVID-19 infection. A retrospective chart review of hospital-
stand the potential consequences of this malarial adaptation ized patients with COVID-19 pneumonia requiring supple-
on the host’s inflammatory responses. mental oxygen found that the cohort with G6PD deficiency
demonstrated prolonged hypoxia and a longer need for me-
Thalassemia. chanical ventilation (19). Outside of COVD-19, no study spe-
Thalassemias are a heterogeneous group of red blood cell dis- cifically addressing whether G6PD deficiency confers a
orders in which the globin-chain units making up normal higher risk for ARDS mortality has been done.
adult hemoglobin (a2b2) are reduced or absent. As a result,
carriers of these mutations show microcytic anemia to vary- Pyruvate kinase deficiency.
ing degrees, and at least in the neonatal period, compensate Pyruvate kinase (PK) deficiency has similarly been linked to
with elevated levels of circulating fetal hemoglobin (c4) (8). resistance against P. falciparum. Pyruvate kinase catalyzes
The prevalence of a- and b-thalassemia have similarly been the rate-limiting step of glycolysis, a process that is crucial to

L172 AJP-Lung Cell Mol Physiol  doi:10.1152/ajplung.00127.2022  www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (189.203.131.199) on March 2, 2024.
RED CELL ADAPTATIONS AND CRITICAL ILLNESS

energy production in nonmitochondrial cells like mature with COVID-19, blood group A was associated with a high risk
erythrocytes. In a study comparing in vitro P. falciparum of acquiring COVID-19, though this did not translate to higher
inoculation of RBCs obtained from homozygous and hetero- rates of intubations in the severely ill patients with COVID-19 in
zygous pyruvate kinase-deficient adults and controls, PK-defi- a separate study (65, 66). The A group is also associated with
cient RBCs showed decreased invasion by P. falciparum (20). acute kidney injury (AKI) risk in critically ill patients with
Furthermore, phagocytosis of ring-stage and mature-stage- trauma or severe sepsis (37). Recent evidence suggests that ABO
infected erythrocytes was increased in samples from patients expression on endothelium drives this association, which raises
deficient in PK, indicating enhanced clearance of parasitized the important question of whether evolutionary adaptations to
cells (20). Additional genotyping studies have indicated a proteins on RBCs have bystander impacts on nonerythroid cells
global geographical codistribution between malaria and high on which they are also expressed (64). Further work will be im-
frequency of PK deficiency (21). portant in determining whether other RBC adaptations indi-
Much like G6PD deficiency, this adaptation results in rectly affect nonerythroid cells.
wide-ranging clinical consequences from mild hemolytic
anemia to complete transfusion dependency. Though high-
CR1
powered studies have yet to be performed to assess the sus- Complement receptor 1 (CR1), a regulatory component of
ceptibility of individuals with PK deficiency to other patho- one of the oldest arms of the innate immune system, the
gens and critical illnesses, the propensity toward anemia complement system, is more abundantly expressed in eryth-
and the increased risk of functional asplenia are likely dele- rocytes than anywhere else in the body (27).
terious consequences accompanying the protection against The hypothesis that CR1 deficiency may be the result of
P. falciparum. selection pressures by malaria-causing species comes from cu-
mulative research showing CR1 to be the host receptor for the
P. falciparum-induced ligand, PfEMP1 (24, 26, 61). CR1 further
PROBABLE ADAPTATIONS TO MALARIA
participates in malaria pathogenicity by aiding in rosetting, the
Beyond the RBC mutations that have been proven to provide process by which parasitized erythrocytes grab on to neighbor-
malaria resistance to their host, there are numerous erythro- ing erythrocytes to spread infection (25). Clinical studies of
cyte mutations that follow similar geographical patterns indi- host susceptibility to malaria show conflicting evidence as to
cating similar evolutionary pressures and have shown some whether CR1 deficiency is beneficial or detrimental, likely at-
early associations with host resistance to infection. tributable to the fact that there are several polymorphisms that
alter receptor expression in different ways. One study showed
ABO Blood Type that a CR1 polymorphism resulted in CR1 deficiency, which
The ABO histo-blood group refers to a family of glycosyl- conferred protection against severe malaria by reducing rosett-
transferases encoded by the ABO gene that catalyze specific car- ing of P. falciparum-infected cells (25). Other studies, however,
bohydrate modifications on glycans and glycoproteins (32). showed that CR1 deficiency polymorphisms are associated
These glycans are present on the surface of RBCs, platelets, en- with an increase in cerebral malaria (26). Supporting these
dothelium, and epithelium. DNA sequencing has placed the de- findings another study demonstrated polymorphisms associ-
velopment of the group O allele before human migration out of ated with higher erythrocyte CR1 were associated with protec-
Africa, and it has been hypothesized that P. falciparum, which tion from severe cerebral malaria (67). Given CR1’s role in
was active in Africa before the early migration of humans and dampening complement activation, one can speculate that
was lethal before or during the human reproductive years, was perhaps the reduction in parasitic load is counterbalanced
the infectious disease driving the emergence the O allele com- with increased inflammation leading to blood-brain barrier
ponent of ABO groups (59). This hypothesis is supported by the breakdown and parasitic access to the CNS.
worldwide distribution of A and O blood groups, the ratio of A recent study suggested that increased function of the al-
which correlates with the global distribution of malaria (59). ternative complement pathway is associated with improved
Several studies have supported the hypothesis that group O survival during critical illness (68). We have demonstrated
is associated with improved outcomes of severe P. falciparum that RBC-bound complement activation products are ele-
infection in both young adults and the fetuses of pregnant vated in both COVID-19 and non-COVID sepsis (69). Yet, loss
women with placental infections compared with group A of CR1 density on erythrocytes in ICU patients with COVID-
(33–36). One study suggested that H antigen may be impor- 19 correlated with disease severity (27). Whether alterations
tant for P. falciparum invasion, however, no other studies in erythrocyte-specific CR1 expression drive sepsis or host
have addressed this hypothesis (60). The prevailing hypothe- responses in critical illness remains unknown. Further stud-
sis is that the infected RBCs expressing the malarial encoded ies will be necessary to elucidate the role of red cell comple-
protein Pfemp contain Duffy binding-like regions that vary in ment regulation in critical illness.
cytoadhesion based on blood group, with group O being the
Toll-Like Receptor 9
least cytoadhesive and blood group A being the most cytoad-
hesive and demonstrating increased rosetting (59, 61, 62). Recognition of nucleic acids is a vital component of the
There is a growing body of work investigating ABO blood innate immune system, which occurs through cytosolic and
groups’ role following trauma and in critical illness (63). A series endosomal Toll-like receptors. Toll-like receptor 9 (TLR9)
of studies demonstrated that the A group, and specifically the has been shown to mediate inflammatory responses to ma-
A1 allele, confers increased risk for ARDS in a cohort of critically larial DNA (70). TLR9 gene polymorphisms are associated
ill patients (64). In a genome-wide association study of patients with symptomatic malaria in Ghanaian children, and TLR9

AJP-Lung Cell Mol Physiol  doi:10.1152/ajplung.00127.2022  www.ajplung.org L173


Downloaded from journals.physiology.org/journal/ajplung (189.203.131.199) on March 2, 2024.
RED CELL ADAPTATIONS AND CRITICAL ILLNESS

has been shown to play an important role in the develop- which is linked to increased resistance to P. falciparum as dis-
ment of protective immunity to P. yoelli, especially in the cussed earlier, also demonstrates a role in ameliorating acute
regulation of pro- and anti-inflammatory cytokines (30, 71). lung injury (ALI) by decreasing IL-1b secretion and neutrophil
TLR9 deficiency has also been shown to confer partial resist- recruitment (78, 79).
ance to lethal infection from P. yoelli, likely due to malaria
parasites requiring TLR9 signaling for immune evasion via Glycophorin
regulatory T cells (31). Glycophorins (GYPs) are another surface protein on
We have identified TLR9 presence on RBCs where it human erythrocytes, which exhibit diverse genetic variation.
plays a role in scavenging cell-free DNA from the circula- Human GYPs are determinants of the major MNS blood
tion during quiescent states (28). During inflammatory group and Gerbich blood group systems, some of which play
states, however, excess DNA binding (including mtDNA as a role as RBC receptors for P. falciparum invasion and other
well as CpG-DNA from pathogens) leads to RBC-TLR9- pathogens (22). GYPA, in particular, interacts with P. falcipa-
driven anemia and innate immune activation (29). RBCs rum protein erythrocyte binding ligand 1 (EBL-1). Recent
loaded with TLR9-bound DNA effectively concentrate and investigation has shown a large structural variant in the
deliver the CpG-containing DNA to tissue macrophages, chromosome 4 region which gives rise to the Dantu glyco-
which in turn phagocytose the RBCs, contributing to sep- phorin variant and is found predominantly in East African
sis-induced anemia. populations and has been associated with a 40% reduction
In a CpG-induced lung injury model in mice, RBC-derived in risk for severe malaria (23). This Dantu variant is believed
TLR9 binds CpG-DNA to ameliorate lung injury (28). to confer protection by increasing RBC surface tension to a
Whether red cell TLR9 alters the host response in human high enough level that merozoite invasion rarely occurs (80).
lung injury remains to be seen, but if found, could indicate The impact, if any, on critical illness survival for those
another mechanism through which RBC adaptation to with Dantu homozygosity remains unclear. However, it is
malaria contributes to its immunomodulatory role to impact tempting to speculate that mutations that alter membrane
host response to critical illness. surface tension may predispose to RBC lysis or thrombosis
thereby driving inflammatory dysregulation, especially in
miRNAs light of RBCs’ role in chemokine trafficking.
RBCs contain abundant micro RNAs (miRNAs), which are
noncoding RNAs that act as critical mediators of posttranscrip- DISCUSSION
tional regulation to enhance or suppress protein translation of
its target. Despite the known abundance of miRNAs in RBCs, The need to better understand the role of RBC adaptions in
the specific roles of these miRNAs remain to be determined. host immunity is driven by the hypothesis that different popu-
Some have posited that erythrocytes may serve as repositories lations may respond differently to modern protocols used in
of miRNAs in the circulatory system, serving not only as treating critical illness. Numerous studies have begun to
markers of disease but as mediators of inflammation (72). address social determinants of health and other host factors
Studies have shown that miRNAs are important for host that may contribute to the host response during critical illness,
defense against intraerythrocytic pathogens like plasmodium whereas factors such as RBC-mediated immunoregulation
species (73). At least two human miRNAs (miRNA-451 and have been overlooked. As reviewed in this paper, we are only
miRNA-233) are more abundant in cells from individuals beginning to uncover the role RBCs play in mediating immune
with sickle cell disease and sickle cell trait than normal red function. Beyond its gas exchange functions, this ubiquitous
blood cells (74). The implication of this abundance in protec- cell traverses the distance of the human body to transport che-
tion against malaria species was proven in in vitro experi- mokines, miRNA, cell-free DNA, and effectively alter, aid, or
ments showing that miRNA-451 can be incorporated into P. dampen the immune response. Selective pressures exerted
falciparum mRNAs where it inhibits mRNA translation and over the millennia in malaria-endemic regions altered the
parasite growth (74). When normal erythrocytes are manipu- makeup of erythrocytes in select individuals, impacting not
lated to overexpress miRNA-451 and miRNA-223, these eryth- only the cargo-carrying capacity but also the type of cargo it
rocytes show reduced parasite growth. Conversely, blocking transports. Recognizing this previously overlooked compart-
the expression of these miRNAs in erythrocytes from those ment in critical illness (along with the selective pressures that
with sickle cell disease and sickle cell trait led to diminished alter the genetic makeup) adds another layer of complexity to
malaria resistance (74). the already heterogeneous population that makes up patients
The numerous mechanisms by which miRNAs play a role with ARDS and sepsis. Yet understanding it better may lend
in regulating immune cell function remain broad and under insight into why some protocolized therapies failed.
investigation. Both intracellular and extracellular miRNAs Take, for example, the multiple studies demonstrating that
have been shown to mediate immune responses. Studies have populations in malaria-endemic regions have not responded to
found that tissue ischemia, like that seen in sepsis, causes protocols developed in cohorts from nonmalaria regions.
release of cellular RNA, including miRNA, into the extracellu- Perhaps the most drastic example of this is the the Fluid
lar space. Once released, it may mediate local and systemic Expansion as Supportive Therapy (FEAST) trial, in which fluid
inflammatory responses via activation of pattern recognition resuscitation strategies that have been widely endorsed by
receptors (75, 76). Alteration of circulating miRNA levels has western-society pediatric life-support training programs were
even been linked to ARDS prognosis, with miRNA-126 show- applied to children with severe febrile illness and impaired per-
ing the potential to predict 28-day mortality (77). miRNA-223, fusion in Uganda, Kenya, and Tanzania, all regions in which

L174 AJP-Lung Cell Mol Physiol  doi:10.1152/ajplung.00127.2022  www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (189.203.131.199) on March 2, 2024.
RED CELL ADAPTATIONS AND CRITICAL ILLNESS

centuries of endemic malaria has exerted its pressures (81). In disease, with blood group A conferring higher risk (66). A
contrast to original research performed on patients in nonen- large, multicenter retrospective chart review conducted from
demic regions, the study found that excess mortality with January 2020 to September 2020 of patients with COVID-19
fluid resuscitation was consistent across all groups, irrespec- found that, compared with Black-identifying individuals
tive of physiological derangement or underlying microbial without sickle cell trait or sickle cell disease, Black-identify-
pathogen. A similar study in an adult population in Zambia in ing individuals with sickle cell disease were at higher risk for
2017 showed increased in-hospital mortality with early resus- hospitalization and the development of pneumonia, even
citation with intravenous fluids and vasopressors compared when controlling for other preexisting conditions (91). RBCs
with usual care (82). In the United States, a subgroup reanaly- obtained during the acute phase of infection from patients
sis of the Fluids and Catheters Treatment Trial (FACTT) trial, with moderate to severe COVID-19 pneumonia (defined as
a study that initially concluded that a restrictive fluid resusci- requiring supplemental oxygen support but not needing in-
tation strategy, was superior to a liberal one in intubated tensive care) were shown to induce endothelial injury via
patients with ARDS. Jolley et al. (83) found that although RBC-derived reactive oxygen species (ROS) and cytokines
there was a decreased 1-yr mortality in black-identifying par- (92). In addition, a small but significant study found that,
ticipants, the conservative fluid strategy actually led to a among hospitalized patients with COVID pneumonia, the
higher 1-yr mortality for white-identifying participants. group with G6PD deficiency was significantly more likely to
Despite limitations, this important observation that there have a worse P:F ratio, spend a longer period of time on
is a heterogeneous treatment response calls for ongoing dis- mechanical ventilation, and have decreased hematocrit
cussions regarding the roles of protocolized versus precision throughout their hospitalization (19). In a small retrospective
medicine. As Dellinger et al. (84) noted, although protocols study, the DARC polymorphism was suggested to be associ-
are often borne out of necessity for quick action in resource- ated with COVID-19 severity infection though the study was
or time-limited settings, and often improve adherence to underpowered to make definitive conclusions (93).
best practice standards, precision-based methods will likely Despite the abovementioned studies, the role of RBC het-
be a critical component of future sepsis treatment. erogeneity remains understudied in critical illness. The col-
The recent work that has been done in defining subgroups lection and biobanking of RBC samples combined with
of patients with different host immune responses in sepsis further translational studies will be necessary to elucidate
lends credence to the benefits of precision medicine that could the immunomodulatory functions of RBCs during critical ill-
enhance well-designed protocols. These distinct phenotypes, ness. Given the accumulating evidence for RBC immune
derived from transcriptomic analysis of leukocytes in periph- function and the recognition that host immune heterogene-
eral blood, have revealed a relatively immunosuppressed ver- ity underlies differential severity to numerous pathogens, it
sus immunocompetent phenotype with differential response is crucial that further investigations promoting precision
to steroid administration—the relatively immunocompetent medicine include large-scale exploration of RBC heterogene-
group showing increased mortality with hydrocortisone use ity. By examining RBC heterogeneity through the most
(85–87). Similarly, patients with acute respiratory distress syn- powerful evolutionary pressure exerted on them—malaria—
drome (ARDS) have been stratified into two subgroups defined we may continue to uncover previously unknown complex-
by levels of inflammatory biomarkers. Phenotype 1 (the hypo- ities of RBC immune function, and therefore host immune
inflammatory subgroup) has lower mortality, more ventilator- diversity. Moreover, given that these adaptations have been
free, and more organ failure-free days compared with the
shaped by pressures that are unequal across geography,
hyperinflammatory Phenotype 2 (88, 89). Much like the tran-
ancestry, and environment, we must account for genetic
scriptionally defined sepsis subgroups described earlier, the
ancestry and environment as potential interacting or con-
importance of stratifying patients in clinical trials by these
founding factors in addition to the socioeconomic disparities
phenotypes has been demonstrated by the use of steroids:
already under investigation. Collectively, investigation of
Sinha et al. (90) recently demonstrated that steroids decreased
adaptations to malaria may provide a framework through
mortality in Phenotype 2 patients with ARDS secondary to
which to better understand this missing piece of host
COVID-19, but not in Phenotype 1 patients. Emerging evidence
immune diversity, in the current pandemic and beyond.
demonstrating RBCs’ immunomodulatory roles coupled with
millennia of selective evolutionary pressures on RBCs call for a
reexamination of RBC heterogeneity as a variable that can ACKNOWLEDGMENTS
modulate the host response to inflammation and infec- The authors acknowledge Kaitlyn Eckart and Julia Yuen for
tion. For example, one critical question would be how administrative assistance. Graphical abstract image created with
would subgroup analysis of the above studies segregate by BioRender.com and published with permission.
RBC adaptations?
The current pandemic provides us with a unique opportu- GRANTS
nity to a single, ubiquitous pathogen through which we can
gain insight into remarkably divergent host responses, This work was supported in part by the NIH Grant R21 AI166813
and by the Penn Center for Precision Medicine at the University of
including those involving RBC heterogeneity. Several of the
Pennsylvania (to N.S.M.).
RBC adaptations discussed in this review have shown signifi-
cant differences in host response to coronavirus infection. A
genome-wide association analysis of almost 2,000 patients DISCLOSURES
with severe COVID-19 (defined by respiratory failure) found No conflicts of interest, financial or otherwise, are declared by
potential involvement of the ABO system in the severity of the authors.

AJP-Lung Cell Mol Physiol  doi:10.1152/ajplung.00127.2022  www.ajplung.org L175


Downloaded from journals.physiology.org/journal/ajplung (189.203.131.199) on March 2, 2024.
RED CELL ADAPTATIONS AND CRITICAL ILLNESS

AUTHOR CONTRIBUTIONS Singhasivanon P, Quintana-Murci L, Sakuntabhai A. Positively


selected G6PD-Mahidol mutation reduces Plasmodium vivax
J.D., L.W., and N.S.M. conceived and designed research; J.D., density in Southeast Asians. Science 326: 1546–1549, 2009.
L.W., and N.S.M. prepared figures; J.D., L.W., and N.S.M. drafted doi:10.1126/science.1178849.
manuscript; J.D., L.W., and N.S.M. edited and revised manuscript; 19. Youssef JG, Zahiruddin F, Youssef G, Padmanabhan S, Ensor J,
J.D., L.W., and N.S.M. approved final version of manuscript. Pingali SR, Zu Y, Sahay S, Iyer SP. G6PD deficiency and severity of
COVID19 pneumonia and acute respiratory distress syndrome: tip of
the iceberg? Ann Hematol 100: 667–673, 2021. doi:10.1007/s00277-
REFERENCES 021-04395-1.
20. Ayi K, Min-Oo G, Serghides L, Crockett M, Kirby-Allen M, Quirt I,
1. Miller LH, Mason SJ, Dvorak JA, McGinniss MH, Rothman IK. Gros P, Kain KC. Pyruvate kinase deficiency and malaria. N Engl J
Erythrocyte receptors for (Plasmodium knowlesi) malaria: Duffy Med 358: 1805–1810, 2008. doi:10.1056/NEJMoa072464.
blood group determinants. Science 189: 561–563, 1975. doi:10.1126/ 21. Machado P, Manco L, Gomes C, Mendes C, Fernandes N, Salome
science.1145213. G, Sitoe L, Chibute S, Langa J, Ribeiro L, Miranda J, Cano J, Pinto
2. Miller LH, Mason SJ, Clyde DF, McGinniss MH. The resistance J, Amorim A, do Rosario VE, Arez AP. Pyruvate kinase deficiency in
factor to Plasmodium vivax in blacks. The Duffy-blood-group ge- sub-Saharan Africa: identification of a highly frequent missense
notype, FyFy. N Engl J Med 295: 302–304, 1976. doi:10.1056/ mutation (G829A;Glu277Lys) and association with malaria. PLoS
NEJM197608052950602. One 7: e47071, 2012. doi:10.1371/journal.pone.0047071.
3. Ferreira A, Marguti I, Bechmann I, Jeney V, Chora A, Palha NR, 22. Amuzu DS, Rockett KA, Leffler EM, Ansah F, Amoako N, Morang’a
Rebelo S, Henri A, Beuzard Y, Soares MP. Sickle hemoglobin con- CM, Hubbart C, Rowlands K, Jeffreys AE, Amenga-Etego LN,
fers tolerance to plasmodium infection. Cell 145: 398–409, 2011. Kwiatkowski DP, Awandare GA. High-throughput genotyping
doi:10.1016/j.cell.2011.03.049. assays for identification of glycophorin B deletion variants in pop-
4. Akide-Ndunge OB, Ayi K, Arese P. The Haldane malaria hypothesis: ulation studies. Exp Biol Med (Maywood) 246: 916–928, 2021.
facts, artifacts, and a prophecy. Redox Rep 8: 311–316, 2003. doi:10.1177/1535370220968545.
doi:10.1179/135100003225002952. 23. Leffler EM, Band G, Busby GBJ, Kivinen K, Le QS, Clarke GM, et al.
5. Allison AC. Protection afforded by sickle-cell trait against subtertian Resistance to malaria through structural variation of red blood cell
malareal infection. Br Med J 1: 290–294, 1954. doi:10.1136/bmj. invasion receptors. Science 356: eaam6393, 2017. doi:10.1126/
1.4857.290. science.aam6393.
6. Mawson AR. The pathogenesis of malaria: a new perspective. Pathog 24. Rowe JA, Moulds JM, Newbold CI, Miller LH. P. falciparum rosetting
Glob Health 107: 122–129, 2013. doi:10.1179/2047773213Y.0000000084. mediated by a parasite-variant erythrocyte membrane protein and
7. Anderson HL, Brodsky IE, Mangalmurti NS. The evolving erythro- complement-receptor 1. Nature 388: 292–295, 1997. doi:10.1038/
cyte: red blood cells as modulators of innate immunity. J Immunol 40888.
201: 1343–1351, 2018. doi:10.4049/jimmunol.1800565. 25. Cockburn IA, Mackinnon MJ, O’Donnell A, Allen SJ, Moulds JM,
8. Shamoun M, Callaghan M. Thalassemia. In: Benign Hematologic Baisor M, Bockarie M, Reeder JC, Rowe JA. A human complement
Disorders in Children: A Clinical Guide, edited by Kamat DM, Frei- receptor 1 polymorphism that reduces Plasmodium falciparum
Jones M. Cham: Springer International Publishing, 2021, p. 91–98. rosetting confers protection against severe malaria. Proc Natl Acad
9. Vento S, Cainelli F, Cesario F. Infections and thalassaemia. Lancet Sci USA 101: 272–277, 2004. doi:10.1073/pnas.0305306101.
Infect Dis 6: 226–233, 2006. doi:10.1016/S1473-3099(06)70437-6. 26. Gandhi M. Complement receptor 1 and the molecular pathogenesis
10. Farmakis D, Giakoumis A, Polymeropoulos E, Aessopos A. Patho- of malaria. Indian J Hum Genet 13: 39–47, 2007. doi:10.4103/0971-
genetic aspects of immune deficiency associated with beta-thalas- 6866.34704.
semia. Med Sci Monit 9: RA19–RA22, 2003. 27. Kisserli A, Schneider N, Audonnet S, Tabary T, Goury A, Cousson
11. Ricerca BM, Di Girolamo A, Rund D. Infections in thalassemia and J, Mahmoudi R, Bani-Sadr F, Kanagaratnam L, Jolly D, Cohen JH.
hemoglobinopathies: focus on therapy-related complications. Acquired decrease of the C3b/C4b receptor (CR1, CD35) and
Mediterr J Hematol Infect Dis 1: e2009028, 2009. doi:10.4084/ increased C4d deposits on erythrocytes from ICU COVID-19
MJHID.2009.028. patients. Immunobiology 226: 152093, 2021. doi:10.1016/j.imbio.
12. Darbonne WC, Rice GC, Mohler MA, Apple T, He bert CA, Valente 2021.152093.
AJ, Baker JB. Red blood cells are a sink for interleukin 8, a leuko- 28. Hotz MJ, Qing D, Shashaty MGS, Zhang P, Faust H, Sondheimer
cyte chemotaxin. J Clin Invest 88: 1362–1369, 1991. doi:10.1172/ N, Rivella S, Worthen GS, Mangalmurti NS. Red blood cells homeo-
JCI115442. statically bind mitochondrial DNA through TLR9 to maintain quies-
13. Horuk R, Chitnis CE, Darbonne WC, Colby TJ, Rybicki A, Hadley cence and to prevent lung injury. Am J Respir Crit Care Med 197:
TJ, Miller LH. A receptor for the malarial parasite Plasmodium vivax: 470–480, 2018. doi:10.1164/rccm.201706-1161OC.
the erythrocyte chemokine receptor. Science 261: 1182–1184, 1993. 29. Lam LKM, Murphy S, Kokkinaki D, Venosa A, Sherrill-Mix S, Casu
doi:10.1126/science.7689250. C, Rivella S, Weiner A, Park J, Shin S, Vaughan AE, Hahn BH,
14. Howes RE, Patil AP, Piel FB, Nyangiri OA, Kabaria CW, Gething Odom John AR, Meyer NJ, Hunter CA, Worthen GS, Mangalmurti
PW, Zimmerman PA, Barnadas C, Beall CM, Gebremedhin A, NS. DNA binding to TLR9 expressed by red blood cells promotes
Me nard D, Williams TN, Weatherall DJ, Hay SI. The global distri- innate immune activation and anemia. Sci Transl Med 13: eabj1008,
bution of the Duffy blood group. Nat Commun 2: 266, 2011. 2021. doi:10.1126/scitranslmed.abj1008.
doi:10.1038/ncomms1265. 30. Gowda NM, Wu X, Gowda DC. TLR9 and MyD88 are crucial for the
15. Mangalmurti NS, Xiong Z, Hulver M, Ranganathan M, Liu XH, development of protective immunity to malaria. J Immunol 188:
Oriss T, Fitzpatrick M, Rubin M, Triulzi D, Choi A, Lee JS. Loss of 5073–5085, 2012. doi:10.4049/jimmunol.1102143.
red cell chemokine scavenging promotes transfusion-related lung 31. Hisaeda H, Tetsutani K, Imai T, Moriya C, Tu L, Hamano S, Duan X,
inflammation. Blood 113: 1158–1166, 2009. doi:10.1182/blood-2008- Chou B, Ishida H, Aramaki A, Shen J, Ishii KJ, Coban C, Akira S,
07-166264. Takeda K, Yasutomo K, Torii M, Himeno K. Malaria parasites require
16. Kangelaris KN, Sapru A, Calfee CS, Liu KD, Pawlikowska L, Witte TLR9 signaling for immune evasion by activating regulatory T cells. J
JS, Vittinghoff E, Zhuo H, Auerbach AD, Ziv E, Matthay MA, Immunol 180: 2496–2503, 2008. doi:10.4049/jimmunol.180.4.2496.
National Heart L, Blood Institute AN; National Heart, Lung, and 32. Dean L. Blood Groups and Red Cell Antigens. Bethesda, MD:
Blood Institute ARDS Network. The association between a Darc National Center for Biotechnology Information (US), 2005.
gene polymorphism and clinical outcomes in African American 33. Pathirana SL, Alles HK, Bandara S, Phone-Kyaw M, Perera MK,
patients with acute lung injury. Chest 141: 1160–1169, 2012. doi:10.1378/ Wickremasinghe AR, Mendis KN, Handunnetti SM. ABO-blood-
chest.11-1766. group types and protection against severe, Plasmodium falciparum
17. Luzzatto L, Ally M, Notaro R. Glucose-6-phosphate dehydrogenase malaria. Ann Trop Med Parasitol 99: 119–124, 2005. doi:10.1179/
deficiency. Blood 136: 1225–1240, 2020. doi:10.1182/blood.2019000944. 136485905X19946.
18. Louicharoen C, Patin E, Paul R, Nuchprayoon I, Witoonpanich 34. Loscertales MP, Brabin BJ. ABO phenotypes and malaria related
B, Peerapittayamongkol C, Casademont I, Sura T, Laird NM, outcomes in mothers and babies in The Gambia: a role for histo-

L176 AJP-Lung Cell Mol Physiol  doi:10.1152/ajplung.00127.2022  www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (189.203.131.199) on March 2, 2024.
RED CELL ADAPTATIONS AND CRITICAL ILLNESS

blood groups in placental malaria? Malar J 5: 72, 2006. doi:10.1186/ 53. Faust HE, Reilly JP, Anderson BJ, Ittner CAG, Forker CM, Zhang P,
1475-2875-5-72. Weaver BA, Holena DN, Lanken PN, Christie JD, Meyer NJ,
35. Lell B, May J, Schmidt-Ott RJ, Lehman LG, Luckner D, Greve B, Mangalmurti NS, Shashaty MGS. Plasma mitochondrial DNA levels
Matousek P, Schmid D, Herbich K, Mockenhaupt FP, Meyer CG, are associated with ARDS in trauma and sepsis patients. Chest 157:
Bienzle U, Kremsner PG. The role of red blood cell polymorphisms 67–76, 2020. doi:10.1016/j.chest.2019.09.028.
in resistance and susceptibility to malaria. Clin Infect Dis 28: 794– 54. Verma A, Huffman JE, Gao L, Minnier J, Wu W-C, Cho K, et al.
799, 1999. doi:10.1086/515193. Association of kidney comorbidities and acute kidney failure with
36. Fischer PR, Boone P. Short report: severe malaria associated with unfavorable outcomes after COVID-19 in individuals with the sickle
blood group. Am J Trop Med Hyg 58: 122–123, 1998. doi:10.4269/ cell trait. JAMA Intern Med 182: 796–804, 2022. doi:10.1001/
ajtmh.1998.58.122. jamainternmed.2022.2141.
37. Reilly JP, Anderson BJ, Mangalmurti NS, Nguyen TD, Holena DN, 55. Flint J, Hill AV, Bowden DK, Oppenheimer SJ, Sill PR, Serjeantson
Wu Q, Nguyen ET, Reilly MP, Lanken PN, Christie JD, Meyer NJ, SW, Bana-Koiri J, Bhatia K, Alpers MP, Boyce AJ. Weatherall DJ,
Shashaty MG. The ABO histo-blood group and AKI in critically ill Clegg JB. High frequencies of alpha-thalassaemia are the result
patients with trauma or sepsis. Clin J Am Soc Nephrol 10: 1911–1920, of natural selection by malaria. Nature 321: 744–750, 1986.
2015. doi:10.2215/CJN.12201214. doi:10.1038/321744a0.
38. Rogers S, Doctor A. Red blood cell dysfunction in critical illness. Crit 56. Weatherall DJ, Clegg JB. Genetic disorders of hemoglobin. Semin
Care Clin 36: 267–292, 2020. doi:10.1016/j.ccc.2019.12.008. Hematol 36: 24–37, 1999.
39. Cutbush M, Mollison PL. The Duffy blood group system. Heredity 57. Allen SJ, O’Donnell A, Alexander ND, Alpers MP, Peto TE, Clegg
(Edinb) 4: 383–389, 1950. doi:10.1038/hdy.1950.31. JB, Weatherall DJ. alpha þ -Thalassemia protects children against
40. Horuk R. The Duffy antigen receptor for chemokines DARC/ACKR1. disease caused by other infections as well as malaria. Proc Natl
Front Immunol 6: 279, 2015. doi:10.3389/fimmu.2015.00279. Acad Sci USA 94: 14736–14741, 1997. doi:10.1073/pnas.94.26.14736.
41. Tournamille C. Molecular basis and structure-activity relationships 58. Luzzi GA, Merry AH, Newbold CI, Marsh K, Pasvol G, Weatherall
of the Duffy blood group antigens: chemokine and Plasmodium DJ. Surface antigen expression on Plasmodium falciparum-infected
vivax receptors. Transfus Clin Biol 7: 497–509, 2000. doi:10.1016/ erythrocytes is modified in alpha- and beta-thalassemia. J Exp Med
s1246-7820(00)80038-5. 173: 785–791, 1991. doi:10.1084/jem.173.4.785.
42. Tournamille C, Filipe A, Wasniowska K, Gane P, Lisowska E, Cartron 59. Cserti CM, Dzik WH. The ABO blood group system and Plasmodium
J-P, Colin Y, Le Van Kim C. Structure–function analysis of the extrac- falciparum malaria. Blood 110: 2250–2258, 2007. doi:10.1182/blood-
ellular domains of the Duffy antigen/receptor for chemokines: charac- 2007-03-077602.
terization of antibody and chemokine binding sites. Br J Haematol 60. Pathak V, Colah R, Ghosh K. Correlation between ’H’ blood group
122: 1014–1023, 2003. doi:10.1046/j.1365-2141.2003.04533.x. antigen and Plasmodium falciparum invasion. Ann Hematol 95:
1067–1075, 2016. doi:10.1007/s00277-016-2663-5.
43. Rot A. Contribution of Duffy antigen to chemokine function. Cytokine
61. Rowe JA, Handel IG, Thera MA, Deans AM, Lyke KE, Kone  A, Diallo
Growth Factor Rev 16: 687–694, 2005. doi:10.1016/j.cytogfr.
DA, Raza A, Kai O, Marsh K, Plowe CV, Doumbo OK, Moulds JM.
2005.05.011.
Blood group O protects against severe Plasmodium falciparum
44. Pruenster M, Mudde L, Bombosi P, Dimitrova S, Zsak M,
malaria through the mechanism of reduced rosetting. Proc Natl Acad
Middleton J, Richmond A, Graham GJ, Segerer S, Nibbs RJ, Rot A.
Sci USA 104: 17471–17476, 2007. doi:10.1073/pnas.0705390104.
The Duffy antigen receptor for chemokines transports chemokines
62. Chen Q, Heddini A, Barragan A, Fernandez V, Pearce SF,
and supports their promigratory activity. Nat Immunol 10: 101–108,
Wahlgren M. The semiconserved head structure of Plasmodium fal-
2009 [Erratum in Nat Immunol 10: 223, 2009]. doi:10.1038/ni.1675.
ciparum erythrocyte membrane protein 1 mediates binding to multi-
45. Lee JS, Wurfel MM, Matute-Bello G, Frevert CW, Rosengart MR,
ple independent host receptors. J Exp Med 192: 1–10, 2000.
Ranganathan M, Wong VW, Holden T, Sutlief S, Richmond A, doi:10.1084/jem.192.1.1.
Peiper S, Martin TR. The Duffy antigen modifies systemic and local 63. Reilly JP, Meyer NJ, Shashaty MG, Feng R, Lanken PN, Gallop R,
tissue chemokine responses following lipopolysaccharide stimulation. Kaplan S, Herlim M, Oz NL, Hiciano I, Campbell A, Holena DN,
J Immunol 177: 8086–8094, 2006. doi:10.4049/jimmunol.177.11.8086. Reilly MP, Christie JD. ABO blood type A is associated with
46. Fukuma N, Akimitsu N, Hamamoto H, Kusuhara H, Sugiyama Y, increased risk of ARDS in whites following both major trauma and
Sekimizu K. A role of the Duffy antigen for the maintenance of severe sepsis. Chest 145: 753–761, 2014. doi:10.1378/chest.13-1962.
plasma chemokine concentrations. Biochem Biophys Res Commun 64. Reilly JP, Meyer NJ, Shashaty MG, Anderson BJ, Ittner C, Dunn
303: 137–139, 2003. doi:10.1016/s0006-291x(03)00293-6. TG, Lim B, Forker C, Bonk MP, Kotloff E, Feng R, Cantu E,
47. Vlaar AP, Juffermans NP. Transfusion-related acute lung injury: a Mangalmurti NS, Calfee CS, Matthay MA, Mikacenic C, Walley KR,
clinical review. Lancet 382: 984–994, 2013. doi:10.1016/S0140-6736 Russell J, Christiani DC, Wurfel MM, Lanken PN, Reilly MP,
(12)62197-7. Christie JD. The ABO histo-blood group, endothelial activation, and
48. Mangla A, Ehsan M, Agarwal N, Maruvada S. Sickle cell anemia. In: acute respiratory distress syndrome risk in critical illness. J Clin
StatPearls. Treasure Island, FL: StatPearls Publishing, 2022. https:// Invest 131: e139700, 2021. doi:10.1172/JCI139700.
www.ncbi.nlm.nih.gov/books/NBK482164/. 65. Zietz M, Zucker J, Tatonetti NP. Associations between blood type
49. Henrici RC, Sautter CL, Bond C, Opoka RO, Namazzi R, Datta D, and COVID-19 infection, intubation, and death. Nat Commun 11:
Ware RE, Conroy AL, John CC. Decreased parasite burden and 5761, 2020. doi:10.1038/s41467-020-19623-x.
altered host response in children with sickle cell anemia and severe 66. Severe Covid-19 GWAS Group, Ellinghaus D, Degenhardt F,
anemia with malaria. Blood Adv 5: 4710–4720, 2021. doi:10.1182/ Bujanda L, Buti M, Albillos A, et al. Genomewide association study
bloodadvances.2021004704. of severe COVID-19 with respiratory failure. N Engl J Med 383:
50. Conran N, Belcher JD. Inflammation in sickle cell disease. Clin 1522–1534, 2020. doi:10.1056/NEJMoa2020283.
Hemorheol Microcirc 68: 263–299, 2018. doi:10.3233/CH-189012. 67. Teeranaipong P, Ohashi J, Patarapotikul J, Kimura R, Nuchnoi P,
51. Tumburu L, Ghosh-Choudhary S, Seifuddin FT, Barbu EA, Yang S, Hananantachai H, Naka I, Putaporntip C, Jongwutiwes S,
Ahmad MM, Wilkins LHW, Tunc I, Sivakumar I, Nichols JS, Dagur Tokunaga K. A functional single-nucleotide polymorphism in the
PK, Yang S, Almeida LEF, Quezado ZMN, Combs CA, Lindberg E, CR1 promoter region contributes to protection against cerebral
Bleck CKE, Zhu J, Shet AS, Chung JH, Pirooznia M, Thein SL. malaria. J Infect Dis 198: 1880–1891, 2008. doi:10.1086/593338.
Circulating mitochondrial DNA is a proinflammatory DAMP in sickle 68. Bain W, Li H, van der Geest R, Moore SR, Olonisakin TF, Ahn B,
cell disease. Blood 137: 3116–3126, 2021 [Erratum in Blood 140: 1327, Papke E, Moghbeli K, DeSensi R, Rapport S, Saul M, Hulver M,
2022]. doi:10.1182/blood.2020009063. Xiong Z, Mallampalli RK, Ray P, Morris A, Ma L, Doi Y, Zhang Y,
52. Caielli S, Cardenas J, de Jesus AA, Baisch J, Walters L, Blanck JP, Kitsios GD, Kulkarni HS, McVerry BJ, Ferreira VP, Nouraie M, Lee
Balasubramanian P, Stagnar C, Ohouo M, Hong S, Nassi L, JS. Increased alternative complement pathway function and improved
Stewart K, Fuller J, Gu J, Banchereau JF, Wright T, Goldbach- survival during critical illness. Am J Respir Crit Care Med 202: 230–
Mansky R, Pascual V. Erythroid mitochondrial retention triggers my- 240, 2020. doi:10.1164/rccm.201910-2083OC.
eloid-dependent type I interferon in human SLE. Cell 184: 4464– 69. Lam LKM, Reilly JP, Rux AH, Murphy SJ, Kuri-Cervantes L,
4479.e19, 2021. doi:10.1016/j.cell.2021.07.021. Weisman AR, Ittner CAG, Pampena MB, Betts MR, Wherry EJ,

AJP-Lung Cell Mol Physiol  doi:10.1152/ajplung.00127.2022  www.ajplung.org L177


Downloaded from journals.physiology.org/journal/ajplung (189.203.131.199) on March 2, 2024.
RED CELL ADAPTATIONS AND CRITICAL ILLNESS

Song W-C, Lambris JD, Meyer NJ, Cines DB, Mangalmurti NS. early resuscitation protocol on in-hospital mortality among adults
Erythrocytes identify complement activation in patients with COVID- with sepsis and hypotension: a randomized clinical trial. JAMA 318:
19. Am J Physiol Lung Cell Mol Physiol 321: L485–L489, 2021. 1233–1240, 2017. doi:10.1001/jama.2017.10913.
doi:10.1152/ajplung.00231.2021. 83. Jolley SE, Hough CL, Clermont G, Hayden D, Hou S, Schoenfeld D,
70. Parroche P, Lauw FN, Goutagny N, Latz E, Monks BG, Visintin A, Smith NL, Thompson BT, Bernard GR, Angus DC, Investigators
Halmen KA, Lamphier M, Olivier M, Bartholomeu DC, Gazzinelli AN; ARDS Network Investigators. Relationship between race and
RT, Golenbock DT. Malaria hemozoin is immunologically inert but the effect of fluids on long-term mortality after acute respiratory dis-
radically enhances innate responses by presenting malaria DNA to tress syndrome. secondary analysis of the National Heart, Lung, and
Toll-like receptor 9. Proc Natl Acad Sci USA 104: 1919–1924, 2007. Blood Institute Fluid and Catheter Treatment Trial. Ann Am Thorac
doi:10.1073/pnas.0608745104. Soc 14: 1443–1449, 2017. doi:10.1513/AnnalsATS.201611-906OC.
71. Omar AH, Yasunami M, Yamazaki A, Shibata H, Ofori MF, 84. Dellinger RP, Levy MM, Schorr CA, Townsend SR. 50 years of
Akanmori BD, Shuaibu MN, Kikuchi M, Hirayama K. Toll-like recep- sepsis investigation/enlightenment among adults-the long and
tor 9 (TLR9) polymorphism associated with symptomatic malaria: a winding road. Crit Care Med 49: 1606–1625, 2021. doi:10.1097/CCM.
cohort study. Malar J 11: 168, 2012. doi:10.1186/1475-2875-11-168. 0000000000005203.
72. Sun L, Yu Y, Niu B, Wang D. Red blood cells as potential reposito- 85. Davenport EE, Burnham KL, Radhakrishnan J, Humburg P, Hutton
ries of MicroRNAs in the circulatory system. Front Genet 11: 442, P, Mills TC, Rautanen A, Gordon AC, Garrard C, Hill AV, Hinds CJ,
2020. doi:10.3389/fgene.2020.00442. Knight JC. Genomic landscape of the individual host response and
73. Duraisingh MT, Lodish HF. Sickle cell microRNAs inhibit the malaria outcomes in sepsis: a prospective cohort study. Lancet Respir Med
parasite. Cell Host Microbe 12: 127–128, 2012. doi:10.1016/j.chom. 4: 259–271, 2016. doi:10.1016/S2213-2600(16)00046-1.
2012.08.001. 86. Antcliffe DB, Burnham KL, Al-Beidh F, Santhakumaran S, Brett SJ,
74. LaMonte G, Philip N, Reardon J, Lacsina JR, Majoros W, Chapman L, Hinds CJ, Ashby D, Knight JC, Gordon AC. Transcriptomic signa-
Thornburg CD, Telen MJ, Ohler U, Nicchitta CV, Haystead T, Chi JT. tures in sepsis and a differential response to steroids. From the
Translocation of sickle cell erythrocyte microRNAs into Plasmodium VANISH randomized trial. Am J Respir Crit Care Med 199: 980–986,
falciparum inhibits parasite translation and contributes to malaria re- 2019 [Erratum in Am J Respir Crit Care Med 206: 1572–1573, 2022].
sistance. Cell Host Microbe 12: 187–199, 2012. doi:10.1016/j.chom. doi:10.1164/rccm.201807-1419OC.
2012.06.007. 87. Wong HR, Hart KW, Lindsell CJ, Sweeney TE. External corrobora-
75. Zou L, He J, Gu L, Shahror RA, Li Y, Cao T, Wang S, Zhu J, Huang tion that corticosteroids may be harmful to septic shock endotype
H, Chen F, Fan X, Wu J, Chao W. Brain innate immune response via A patients. Crit Care Med 49: e98–e101, 2021. doi:10.1097/CCM.
miRNA-TLR7 sensing in polymicrobial sepsis. Brain Behav Immun 0000000000004709.
100: 10–24, 2022. doi:10.1016/j.bbi.2021.11.007. 88. Famous KR, Delucchi K, Ware LB, Kangelaris KN, Liu KD,
76. Xu J, Feng Y, Jeyaram A, Jay SM, Zou L, Chao W. Circulating Thompson BT, Calfee CS; ARDS Network. Acute respiratory dis-
plasma extracellular vesicles from septic mice induce inflammation tress syndrome subphenotypes respond differently to randomized
via microRNA- and TLR7-dependent mechanisms. J Immunol 201: fluid management strategy. Am J Respir Crit Care Med 195: 331–
3392–3400, 2018. doi:10.4049/jimmunol.1801008. 338, 2017. doi:10.1164/rccm.201603-0645OC.
77. Wu X, Wu C, Gu W, Ji H, Zhu L. Serum exosomal MicroRNAs predict 89. Sinha P, Delucchi KL, Thompson BT, McAuley DF, Matthay MA,
acute respiratory distress syndrome events in patients with severe Calfee CS; NHLBI ARDS Network. Latent class analysis of ARDS
community-acquired pneumonia. Biomed Res Int 2019: 3612020, subphenotypes: a secondary analysis of the statins for acutely
2019. doi:10.1155/2019/3612020. injured lungs from sepsis (SAILS) study. Intensive Care Med 44:
78. Feng Z, Qi S, Zhang Y, Qi Z, Yan L, Zhou J, He F, Li Q, Yang Y, 1859–1869, 2018. doi:10.1007/s00134-018-5378-3.
Chen Q, Xiao S, Li Q, Chen Y, Zhang Y. Ly6G þ neutrophil-derived 90. Sinha P, Furfaro D, Cummings MJ, Abrams D, Delucchi K, Maddali
miR-223 inhibits the NLRP3 inflammasome in mitochondrial DAMP- MV, He J, Thompson A, Murn M, Fountain J, Rosen A, Robbins-
induced acute lung injury. Cell Death Dis 8: e3170–e3170, 2017. Juarez SY, Adan MA, Satish T, Madhavan M, Gupta A, Lyashchenko
doi:10.1038/cddis.2017.549. AK, Agerstrand C, Yip NH, Burkart KM, Beitler JR, Baldwin MR,
79. Jiang Z-F, Zhang L, Shen J. MicroRNA: potential biomarker and tar- Calfee CS, Brodie D, O’Donnell MR. Latent class analysis reveals
get of therapy in acute lung injury. Hum Exp Toxicol 39: 1429–1442, COVID-19-related acute respiratory distress syndrome subgroups with
2020. doi:10.1177/0960327120926254. differential responses to corticosteroids. Am J Respir Crit Care Med
80. Kariuki SN, Marin-Menendez A, Introini V, Ravenhill BJ, Lin YC, 204: 1274–1285, 2021. doi:10.1164/rccm.202105-1302OC.
Macharia A, Makale J, Tendwa M, Nyamu W, Kotar J, Carrasquilla 91. Singh A, Brandow AM, Panepinto JA. COVID-19 in individuals with
M, Rowe JA, Rockett K, Kwiatkowski D, Weekes MP, Cicuta P, sickle cell disease/trait compared with other Black individuals. Blood
Williams TN, Rayner JC. Red blood cell tension protects against Adv 5: 1915–1921, 2021 [Erratum in Blood Adv 6: 2535, 2022].
severe malaria in the Dantu blood group. Nature 585: 579–583, doi:10.1182/bloodadvances.2020003741.
2020. doi:10.1038/s41586-020-2726-6. 92. Mahdi A, Collado A, Tengbom J, Jiao T, Wodaje T, Johansson N,
81. Maitland K, Kiguli S, Opoka RO, Engoru C, Olupot-Olupot P, Akech Farnebo F, Fa €rnert A, Yang J, Lundberg Jon O, Zhou Z, Pernow J.
SO, Nyeko R, Mtove G, Reyburn H, Lang T, Brent B, Evans JA, Erythrocytes induce vascular dysfunction in COVID-19. JACC Basic
Tibenderana JK, Crawley J, Russell EC, Levin M, Babiker AG, Gibb Transl Sci 7: 193–204, 2022. doi:10.1016/j.jacbts.2021.12.003.
DM, Group FT; FEAST Trial Group. Mortality after fluid bolus in 93. Conrado MCAV, Dezan MR, Oliveira VB, Ziza KC, Fanciscani T,
African children with severe infection. N Engl J Med 364: 2483– Rocha V, Mendrone-Júnior A, Dinardo CL. Association between
2495, 2011. doi:10.1056/NEJMoa1101549. FY02N.01 and the severity of COVID-19: initial observations.
82. Andrews B, Semler MW, Muchemwa L, Kelly P, Lakhi S, Hematol Transfus Cell Ther 44: 213–217, 2022. doi:10.1016/j.
Heimburger DC, Mabula C, Bwalya M, Bernard GR. Effect of an htct.2022.01.002.

L178 AJP-Lung Cell Mol Physiol  doi:10.1152/ajplung.00127.2022  www.ajplung.org


Downloaded from journals.physiology.org/journal/ajplung (189.203.131.199) on March 2, 2024.

You might also like