Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Dec 10.
Published in final edited form as: Am J Hematol. 2017 Apr 29;92(6):569–582. doi: 10.1002/ajh.24750

The Role of Carbon Monoxide and Heme Oxygenase in the Prevention of Sickle Cell Disease Vaso-Occlusive Crises

Edward Gomperts 1, John D Belcher 2, Leo Otterbein 3, Thomas D Coates 4, John Wood 5, Brett E Skolnick 6, Howard Levy 7, Gregory M Vercellotti 8
PMCID: PMC5723421  NIHMSID: NIHMS923945  PMID: 28378932

1.0. Overview of Sickle Cell Disease

Sickle cell disease (SCD) is a painful, lifelong hemoglobinopathy with substantial morbidities and premature mortality. It is inherited as a point mutation in the hemoglobin (Hb) beta-globin gene where glutamic acid at position 6 is substituted by valine. This mutation provides resistance to malaria, but, in the homozygous form, results in SCD, which is characterized by anemia and painful vaso-occlusive crises (VOCs). The change in molecular structure allows Hb in the deoxygenated state to form polymers that promote Hb autoxidation, red blood cell (RBC) membrane damage, decreased RBC deformability, intravascular and extravascular hemolysis, inflammation, vaso-occlusion, and ultimately organ injury1.

Intravascular hemolysis results in the release of free Hb into plasma. In normal individuals, free Hb is rapidly bound to circulating haptoglobin with high affinity (Kd = ~10−12 M) and carried to CD163 receptors on macrophages for endocytosis and degradation2,3,4. However in SCD patients, the continuous release of Hb from hemolyzed sickle RBCs depletes the plasma haptoglobin resulting in free Hb circulating in plasma5. Free Hb in the vasculature rapidly quenches the vasodilator nitric oxide (NO) resulting in vasospasm6,7. In the pro-oxidative SCD vasculature, the reaction of NO and other oxidants with ferrous Hb oxidizes the heme iron from ferrous Hb to ferric Hb (metHb). MetHb is unstable and releases hemin into the vascular space and it is hemin release that initiates the pathophysiological cascade ultimately resulting in the vascular damage underpinning the widespread tissue and organ damage that is the hallmark of this devastating disease8,9,10.

In normal individuals, the released hemin is rapidly bound to circulating hemopexin with high affinity (Kd< 10−13 M) and carried to CD91 (LRP1) receptors on hepatocytes for endocytosis and degradation11,12,13. However, in SCD patients the continuous formation of metHb in plasma and release of hemin depletes plasma hemopexin levels, in a similar manner to hemoglobin depleting haptoglobin, resulting in free hemin bound to lower affinity RBC-derived microparticles, albumin and lipoproteins14,15,16,17. Hemin is released continuously from these carriers to cell membranes of circulating immune cells, platelets and endothelial cells in the vessel wall18,14. Hemin in the cell membrane is cytotoxic, promotes oxidative stress, and activates innate immune toll-like receptor 4 (TLR4) signaling initiating activation of monocytes and endothelium8,19,20,21,22,23. The resulting inflammatory cascades include degranulation of endothelial Weibel-Palade bodies, enzymatically-derived oxidant production, NF-κB-driven inflammasome activation, pro-inflammatory cytokine production, and adhesion molecule expression. Hemin, cytokines, lipopolysaccharide (LPS), infections, and the microbiome can all promote pro-inflammatory responses leading to adhesion of sickle RBCs, leukocytes, and platelets to the endothelium and the formation of multicellular aggregates leading to vaso-occlusion8,24,25,26,27,28. The subsequent re-opening of occluded vessels promotes ischemia-reperfusion pathophysiology, inflammatory pain, and tissue injury that amplify the innate immune inflammatory responses29,30,31.

SCD manifests clinically through a variety of severe morbidities, including stroke, auto-splenectomy with risk of resultant overwhelming infection, pulmonary hypertension, acute chest syndrome, infarction of organs, priapism, leg ulcers, and renal injury32. According to the Centers for Disease Control (CDC), the median survival among adults in North America is approximately 42 years although a higher median survival rate was reported in an adult-only study in patients with SCD in the United Kingdom33. In addition to the impact on survival, the ravages of this microvascular occlusive disease severely and progressively impact the quality of life from infancy through adulthood34.

Although sickle cell trait has been shown to have evolved in equatorial Africa, individuals with sickle trait have migrated to most major geographic regions of the world35. It is estimated that SCD affects approximately 100,000 Americans36, as well as approximately 37,000 patients in the EU37, and millions of patients globally. The prevalence of the disease is increasing as infant survival rises worldwide35. The number of children born with SCD is expected to exceed 14 million worldwide in the next 40 years, with about 79% of these children being born in sub-Saharan Africa in 2010 where the mortality is highest under 5 years of age 38.

A substantial medical need exists to reduce VOCs, also termed sickle cell crises and the consequent morbidities and mortality. Hydroxyurea remains the only therapeutic approved for the prevention of VOCs in SCD. Hydroxyurea reduces, but does not eliminate the incidence of VOC’s or the morbidities and mortality of SCD. In response to the unmet medical need, there are new therapeutics in development that target a variety of mechanisms, each with its own unique potential risks and benefits39. This review is focused on carbon monoxide (CO), as this small molecule gasotransmitter can impart potent salutary medical benefits based upon specific mechanisms of action at non-toxic low doses. We discuss the promising data that have emerged showing the benefits of CO and the efforts underway to develop it as a potent therapeutic.

2.0. Overview of the Heme Oxygenase/Carbon Monoxide Pathway

The discovery of nitric oxide (NO) gas in 1987 revealed the novel concept that endogenous production of a gas as a signal transmitter can impart diverse and critical systemic physiologic effects across a wide spectrum of biological and pathological processes. Unlike NO which is highly reactive, CO is stable. CO binds to cellular heme moieties and modulates the activity of heme-containing enzymes including mitochondrial oxidases, guanylate cyclase, nitric oxide synthase and NADPH oxidase40,41. Ultimately, the target for CO will depend on the cell type and the expression of hemoproteins that are present in the cell. For example, guanylate cyclase is highly present in vascular smooth muscle cells, but very low in endothelial cells.

CO is generated endogenously when heme is degraded by the heme oxygenase (HO) enzymes. Most, if not all, endogenous CO is generated by HO degradation of heme. These enzymes generate CO in the cytosol. In recent years, interest in HO has grown beyond its role as a metabolic enzyme that degrades heme with research exploding across multiple scientific disciplines. HO is now classified as an anti-inflammatory cytoprotective enzyme that generates bioactive CO that mimics many of the effects of HO and has been described as an anti-inflammatory gasotransmitter. In addition, heme degradation by HO produces iron that is stored in ferritin and the bile pigment biliverdin that is metabolized to bilirubin by biliverdin reductase (Figure 1).

Figure 1.

Figure 1

Heme degradation releases bioactive products

Intravascular hemolysis of RBCs with the resultant free Hb and heme within the circulation is a normally occurring event, albeit occurring at a low rate. Normally, haptoglobin and hemopexin scavenge circulating Hb and heme, respectively, and deliver them to macrophages and hepatocytes for processing by HO. It is when these systems are overwhelmed that pathology such as those of SCD result. Free heme in the presence of reactive oxygen species produced by activated granulocytes is cytotoxic to endothelium18. Heme uptake by endothelium synergizes with polymorphonuclear granulocyte-mediated damage42. In addition, heme activates innate immune TLR4 signaling on endothelium, leukocytes and platelets leading to vascular cell activation that includes oxidant production, NF-κB activation, adhesion molecule expression, pro-inflammatory cytokine production, and necroptosis/apoptosis. In many instances these reactions amplify tissue injury caused by initiating events such as trauma, infection, and/or hemolysis. Detoxification of free heme by HO and the mitigation of heme-induced innate immune responses are critical for survival.

There are two HO enzymes, HO-1 and HO-2, that degrade heme into equimolar quantities of iron, biliverdin and CO (Figure 1,43,44). The isoforms differ in their structure, location and biological role. HO-1 is highly inducible and found in all cells and is upregulated by a variety of agents including heme, pathogens, ischemia/reperfusion injury (IRI), oxidants, and, in certain instances, CO itself45,46,47,48,49,50,51. HO-2 is constitutively expressed primarily in the brain, vasculature, and testes where it is involved in neurotransmission, vasomotor tone, and spermatogenesis52,53. HO-1 and CO are recognized as critically protective due to their ability to modulate inflammation and apoptosis40,54,55. At low doses, equivalent to that generated by HO-1, CO has been shown in a substantial number of research observations to induce HO-1 itself, as well as modulate a wide spectrum of genes linked to inhibition of oxidative stress, inflammation, and apoptosis, including Nrf2, NF-κB, STAT3, CREBH and various MAP kinases51,54,56,57,58,59,60,61. Importantly, these effects of CO have been documented across a variety of CO administration approaches, including increasing endogenous production of CO43, inhalation of CO gas, and intravenous administration of CO in the form of CO attached to a transition metal, such as molybdenum and ruthenium (CO Releasing Molecules [CORMs]), or even iron as a CO-saturated Hb40,51.

CO is a tasteless, odorless, non-corrosive gas that exists as a stable diatomic molecule consisting of one carbon and one oxygen atom. It is physiologically present in all species that utilize heme protein. In healthy humans basal levels of CO in blood range between 0.5 and 1.5% carbon-monoxy hemoglobin (COHb)62. CO binds competitively with oxygen for Hb, with an affinity for Hb 200–250 times that of oxygen. In addition to decreasing the oxygen carrying capacity of Hb, the binding of CO to Hb locks Hb in the R state, increasing the affinity of Hb for oxygen and thereby shifting the oxygen dissociation curve to the left. In addition to Hb, CO also binds to myoglobin and a variety of intracellular hemeproteins including cytochromes and other metallo-enzymes. Exogenous CO is generated during the combustion of fossil fuels, or emitted from the oceans or volcanoes where it is found in large quantities. CO is rapidly taken into the body by inhalation from the atmosphere. It is readily absorbed and excreted through the lungs and rapidly distributed by the blood stream essentially unchanged except for a very low percentage that is converted to CO262. In the tissues a portion of CO offloads from Hb, much like O2, where it can influence cellular function.

The toxicological effects of CO have been well studied. The half-life of CO in sedentary adults at sea level is 4 to 5 hours62. Blood COHb levels are considered a reliable biological marker of the dose or exposure of CO an individual has received. Levels of COHb above 20–30% are associated with adverse symptoms such as headaches and dizziness. Mortality is documented with levels above 50%62,63,64. Levels of COHb lower than 15% are well tolerated for both short and prolonged time periods, as indicated in the epidemiological and clinical literature62,63,64. It is reported that exertional capacity is reduced when COHb is above 5% and individuals with advanced cardiovascular disease are at risk of symptoms with exertion when levels are above 3%62. Headache and nausea have been reported when COHb is raised above 5%, yet rigorous Phase I safety trials have documented that COHb of up to 13.9% showed no adverse effects (see Safety Studies of CO below) and ongoing Phase 2 trials have documented few related adverse events. It should also be noted that there is controversy and debate as to reproducible physiologic effects of CO on the body and the accompanying symptoms that result65.

2.1. Carbon Monoxide as a Gasotransmitter

In contrast to the established CO toxicity demonstrated at high COHb levels, low-dose CO is now accepted as a potent gasotransmitter, capable of regulating a host of physiologic and therapeutic processes40,66. In recent years, research has focused extensively on the mechanisms underlying how CO imparts its salutary effects. The large body of generated data clearly shows that CO provides cytoprotection in models of sterile inflammation including organ transplantation67, acute lung injury68, SCD, stroke69, and other conditions43,70,71. In each instance the protection observed was associated with modulation of the innate and adaptive immune response and restoration of tissue and organ function. One underlying theme is that CO at low concentrations abrogates IRI providing cytoprotection as demonstrated in an IRI lung injury model72. Research has shown the paradoxical effect that CO increased tissue oxygen availability by displacing O2 within intracellular compartments72. In the lung, CO-mediated protection in wild-type mice was abrogated in mice null for the gene encoding plasminogen activator inhibitor (Pai1−/−) mice. These data established a fundamental link between CO, hemostasis and the prevention of IRI based on the ability of CO to derepress the fibrinolytic axis. CO has been shown to be effective both prophylactically and therapeutically even when started at the peak of pathology. In short, CO seems to befit the need of the tissue to restore normal function. This has been defined as homeo-’dynamic’40.

3.0. Overview of Potential Carbon Monoxide Actions in Sickle Cell Disease

How could CO modulate SCD? Exogenous CO may potentially affect SCD through six distinct mechanisms of action (Table 1). First, CO could affect HbS polymerization. CO can bind to HbS polymers and enhance their melting which minimizes their persistence in red cells within the circulation, and their ability to rapidly seed additional polymers during transit in hypoxic tissues. These polymers may contribute to abnormal rheological flow and ultimately vaso-occlusion73. However, the equilibrium solubility of HbS with CO is linearly proportional to the amount of bound CO74. Thus, very high concentrations of CO may be required to have a clinical benefit in SCD through this mechanism. Yet, Sirs showed a reduction of sickle RBCs from 10.2% to 3.9% with a COHb level of 4% in blood from a single patient75, and there have been reports on studies of CO effects on HbS polymerization, with one showing in vitro binding of CO to sickle polymers73 and a second study utilizing a microfluidic device showing concentrations of CO as low as 0.1% preventing occlusion when perfused with blood from SCD individuals76. The melting of polymers may be a key event minimizing the persistence of these polymers in the circulation and as a consequence, limiting these acting as a seed for rapid polymerization in hypoxic tissue73. Further, given the high affinity of CO for Hb, it is a logical supposition that the impact of CO on polymerization could limit sickle-associated clinical effects, but the extent and duration of these effect in vivo have received little focus. Second, CO inhibits the Ca2+-permeable cation conductance channel or P-sickle, thereby inhibiting sickle RBC dehydration77. It has been postulated that an increase in RBC hydration could decrease the rate of HbS polymer formation.

Table 1.

Potential CO targets and mechanisms of action in SCD.

Potential Mechanism Potential Effect Potential Pathways/Targets/Markers
1. Anti-polymerization7376 CO binds to HbS, modulating HbS polymerization Ferrous HbS
2. Sickle red blood cell hydration77 Inhibition of Ca2+-permeable cation conductance Psickle
3. Anti-oxidative7884 Decreased oxidative stress Decreased HbS oxidation, inhibition of NADPH oxidase, and activation Nrf-2-responsive genes including HO-1 and anti-oxidant enzymes.
4. Anti-inflammatory51,54,5661,104115 Modulation of pro- and anti-inflammatory mediators Activation of Nrf-2-responsive genes including HO-1 and anti-inflammatory proteins; decreased NF-κB activation leading to decreased VCAM-1, ICAM-1, P-selectin, and pro-inflammatory cytokines; increased nuclear phospho-p38 MAPK and phospho-Akt; decreased innate immune TLR4 signaling and cell membrane expression; and decreased inflammasome activation
5. Vasodilation93,94 Increased cGMP Guanylate cyclase and indirectly by nitric oxide synthase (NOS)
6. Anti-apoptotic97100,151153 Decreased mitochondrial damage and cell death Cytochrome C release, caspase activation, and mitochondrial membrane potential

Third, CO may have anti-oxidative effects by inhibiting the oxidation of ferrous HbS to ferric HbS through its ligation to heme and thereby preventing heme release from ferric HbS78,79. CO may also inhibit superoxide production by NADPH oxidase 2 (NOX2) through CO binding to heme groups on the gp91phox subunit80. In addition, CO activates Nrf2-responsive anti-oxidant genes including enzymes involved in glutathione production and regeneration, NADPH production, and iron sequestration such as HO-1 and ferritin.

Fourth, CO has remarkable anti-inflammatory properties through the activation of Nrf2-responsive genes leading to HO-1 upregulation and NF-κB downregulation51. A wide spectrum of genes are modulated by CO including Nrf2, NF-κB, STAT3, CREBH and various MAP kinases51,54,56,57,58,59,60,61. CO has been shown to modulate inflammation by increasing HIF1α and PPARγ signaling, largely dependent on the ability of CO to generate reactive oxygen species, generated in large part by the mitochondria81,82,83,84. Clues to the protective effect of CO are delineated in the work of Ferreira et al showing that mice with HbS are resistant to malaria infection, due in part to the induction of HO-1 and its products including CO85. HO-1 is highly inducible by CO and both HO-1 and CO modulate inflammatory responses51,54,56,57,58,59,60,61. HO-1 and CO-mediated inhibition of NF-κB activation reduces adhesion molecule and pro-inflammatory cytokine expression, which inhibits vaso-occlusion in SCD mouse models. Other anti-inflammatory effects include increases in phospho-p38 MAPK and phospho-Akt as well as decreases in innate immune TLR4 signaling and TLR4 trafficking to the cell membrane surface86,87,88. CO also inhibits NLRP3 inflammasome activation89,90,91,92. Fifth, NO bioavailability is decreased in SCD. However, CO can substitute for NO binding to the heme protein guanylate cyclase and increase cGMP production leading to vasodilation of blood vessels93,94. CO has been shown to have a relationship with NO in the vasculature where CO increases endothelial cell repair after injury and can induce apoptosis of hyperproliferative vascular smooth muscle cells that cause pulmonary hypertension. In each instance, CO increases NO bioavailability that is involved in the observed benefits95,96. Sixth, CO has anti-apoptotic activity by preventing mitochondrial depolarization, inhibiting cytochrome C release, suppressing p53 expression, and inhibiting caspase activation97,98,99,100.

Although the potential efficacy of CO in SCD may rely upon all of these mechanisms of action, the exact dosing regimen of CO and resulting COHb concentration to maximize efficacy and ensure safety, will depend on the interplay of these mechanisms in the clinical setting. Currently available data indicates that given the relatively short half-life of CO, the prevention of polymerization through CO binding to HbS (Mechanism 1 above) and the vasodilatory effects of CO (Mechanism 5) are anticipated to require more frequent and higher doses, while the anti-inflammatory and anti-apoptotic actions (Mechanisms 4 and 6, respectively) will likely require less frequent and lower doses. Preclinical data suggests that, in transgenic SCD mice, smaller, daily doses with peak COHb levels at 2% to 6.5% are effective in down-regulating the pathophysiological mechanisms that are associated with SCD51,101.

3.1. Reported Studies of CO in Sickle Cell Disease

A substantial number of studies, including cell based models and in vivo in animals and humans have been conducted supporting the potential efficacy, safety, and bioavailability of CO in SCD using a variety of delivery mechanisms, including inhaled and parenteral administration.

3.1.1. Preclinical In Vitro Sickle Cell Disease Studies

A number of in vitro studies have demonstrated the potential for CO to directly limit the sickling phenomenon through binding to HbS. As early as 1963, it was shown that the presence of CO reduces the proportion of sickled cells in vitro75. A more recent study in a microfluidic device confirmed that a small CO concentration prevented occlusion, even without the presence of oxygen76. It has also been demonstrated that CO binds to and melts HbS polymers73. In addition, Bains and colleagues demonstrated that HO-1 induction, exogenous biliverdin or CO administration by CORMs markedly decreased adhesion of RBC’s from sickle cell patients to cultured endothelial cells102.

3.1.2. Preclinical In Vivo Sickle Cell Disease Efficacy Studies

Data from four studies in four different transgenic SCD mouse models demonstrate that modulation of the HO-1/CO pathway is extremely effective in thwarting SCD pathophysiology and suggests low dose CO as a novel treatment approach. A seminal study by Belcher and coworkers first showed the beneficial effects of both the up-regulation of HO-1 and administration of CO in two SCD mouse models103. Their study demonstrated that the up-regulation of HO-1 modulates the pathobiology of SCD. Overexpression of HO-1 in both S+S-Antilles and Berkeley SCD transgenic mice led to significantly reduced hypoxia/reoxygenation-induced vascular stasis. This protective effect of HO-1 was associated with the mitigation of inflammation, including the reduction in leukocyte-endothelium interactions and the inhibition of NF-κB, VCAM-1, and ICAM-1 expression. In contrast, the inhibition of HO-1 activity was shown to exacerbate vascular stasis. Most importantly, these anti-inflammatory and anti-vaso-occlusive effects of HO-1 were nearly identical in sickle mice inhaling low concentrations (250 parts per million [ppm]) of CO gas for 1 hour over 3 consecutive days).

A parallel study confirmed these effects of HO-1 in limiting vaso-occlusion and inflammation in SCD mice. In this study, livers of sickle mice overexpressing wild type HO-1, but not an enzymatically inactive HO-1, showed marked activation of the phospho-p38 MAPK and phospho-Akt cell signaling pathways, reduced levels of NF-κB p65 in liver nuclear extracts, and decreased sVCAM-1 in serum104. Similarly, hypoxia-induced vascular stasis was only inhibited in sickle mice overexpressing enzymatically active HO-1. Vascular stasis was inhibited in the venules of dorsal skin despite the absence of the HO-1 transgene in the skin, indicating that overexpression of the wild type HO-1 transgene in the livers of sickle mice had distal effects on the vasculature suggesting a mediator that could be transported and exert effects remotely.

This work was extended further investigating the anti-inflammatory effects of CO in S+S-Antilles SCD mice101. In this study, treatment of SCD mice with 25 or 250 ppm of inhaled CO for 1 hour per day, 3 days per week for 8 to 10 weeks showed decreased vascular inflammation and end-organ pathology. Inhaled CO treatment significantly reduced total white blood cell, neutrophil and lymphocyte counts. Moreover, reduced staining for myeloid and lymphoid markers in the bone marrow was observed, and bone marrow exhibited a significant decrease in colony-forming units by granulocyte-macrophage colony-forming cell assays. Anti-inflammatory signaling pathways including phospho-Akt and phospho-p38 MAPK were also markedly increased in the livers of CO-treated mice. Importantly, treated sickle mice had a significant reduction in liver parenchymal necrosis, reflecting the anti-inflammatory and anti-vaso-occlusive benefits of CO administration, and these effects were seen at a peak COHb level of 2% and at higher levels.

A follow-on study demonstrated that the protective effects of CO extended to two additional SCD transgenic mouse models using a PEGylated COHb molecule (MP4CO) for intravenous CO administration51. This effect was not seen with the oxygenated molecule MP4OX. Similar to the studies described above, the administration of CO in the NY1DD transgenic SCD mouse model markedly induced HO-1 activity and inhibited NF-κB activation and hypoxia/reoxygenation-induced microvascular stasis. Importantly, CO administration also upregulated nuclear Nrf2, an important transcriptional regulator of HO-1 and other cytoprotective antioxidant genes. Additionally, in a heterozygous HbAS-Townes sickle trait mouse model, administration of CO significantly reduced mortality in a model of acute lung injury and cardiac dysfunction. CO administration decreased NF-κB activation, P-selectin and von Willebrand Factor expression on blood vessels, indicating anti-inflammatory and anti-adhesive effects. Importantly, these effects were seen at peak COHb levels of 6.5%.

3.1.3. Sickle Cell Disease Clinical Efficacy and Epidemiologic Studies

Data from three independent studies provide preliminary clinical evidence for the COmediated prevention of VOC in SCD patients. An early clinical study by Sirs first demonstrated that a 30 minute administration of inhaled CO to a SCD patient producing an estimated COHb concentration of 4%, resulted in a 60% reduction of sickled RBCs in the peripheral blood75. Beutler and colleagues extended this work, demonstrating that the administration of inhaled CO to two SCD adult patients increased Cr51-tagged RBC survival, suggesting that the rheologic properties of sickle cells were favorably influenced105. Importantly, although these two studies comprise a very limited number of patients, the Sirs study suggests the possibility of a relatively low minimum effective dose, which clearly will need to be studied in formal clinical trials. In addition, an epidemiologic study carried out in London showed a significantly lower hospital admission rate for VOC on days when higher ambient atmospheric levels of CO were recorded106. Importantly, this significantly lower hospital admission rate was not related to the other five atmospheric pollutants that were investigated.

3.1.4. Safety Studies of CO

The clinical safety of CO in SCD depends on the target dose and COHb levels. As discussed above, a target dose resulting in COHb levels as low as 2% has been reported to be efficacious in SCD in preclinical studies, and a COHb of approximately 4% has been reported to significantly reduce sickled RBCs in a clinical experience, suggesting a target range of <10% COHb as appropriate to evaluate for efficacy.

At the time of writing, a review of clinicaltrials.gov show there are 8 completed and 10 on-going clinical studies testing a variety of CO-delivery modalities (but not oral) (Table 2). Thus 18 studies with a variety of sponsors have Phase 1 and Phase 2 clinical investigations under regulatory guidance, providing strong support for the perception of safety at targeted CO-Hb levels for Phase 1 and Phase 2 clinical investigation.

Table 2.

Completed Phase 1 or Phase 2 clinical studies* supported by a number of sponsors and employing a variety of CO-administration mechanisms

Clinical Trial Description/Indication CO Product
Phase 1 Single Dose Safety (Normal Volunteers) Inhaled gas107
Phase 1 Repeat Dose (10 Days) Safety (Normal Volunteers) Inhaled gas107
Phase 1 Single Dose Safety (Normal Volunteers) CORM (Sanguinate)110
Phase 1b Repeat Dose Safety (Sickle Cell Disease) CORM (Sanguinate)109
Phase 1b Repeat Dose Safety (Sickle Cell Disease) CORM (MP4CO)108
Phase 2 Chronic Obstructive Pulmonary Disorder Inhaled gas111
Phase 2 Experimental Endotoxemia Inhaled gas112
Phase 2 Mitochondrial Biogenesis (skeletal muscle) Inhaled gas113

Two successful Phase 1 clinical trials in normal volunteers have been conducted with inhaled CO gas, demonstrating the safety and tolerability of CO up to a peak COHb level of 13.9%107. In addition, two Phase 1b clinical safety studies conducted with CO using either intravenous MP4CO or Sanguinate, demonstrated safety and tolerability in SCD patients in all dose groups, with a COHb increase over baseline of 2% for MP4CO108,109. The potential for therapeutic development of CO is demonstrated by four additional Phase 1 and 2 clinical studies with inhaled and Hb-delivered CO conducted in non-SCD indications110,111,112,113.

Despite these supportive studies, the safety of CO at concentrations at or below COHb of 10% in SCD patients is based on limited data. Individuals with SCD may have underlying SCD-associated end-organ disease that may be negatively impacted by CO administration, which may result from less oxygen to tissues. It is also possible, but unlikely given the current data, that CO may actually increase HbS polymerization. Given the current limited information on the impact of CO in individuals with SCD, careful study in SCD patients is warranted.

4.0. CO and Neuronal Injury

CO has been shown to be of benefit in a number of animal models of organ ischemia, including brain, kidney, heart, liver, and gut ischemia. In SCD there are major concerns around cerebral ischemia, as cerebrovascular occlusive events (CVOE) in patients with SCD are common and are major clinical events that uniformly determine adverse outcomes114. These events can manifest across the age spectrum as overt strokes, but cerebral infarcts can also be silent, with the cumulative events over time resulting in chronic cerebral ischemia, cognitive impairment, and poor quality of life115,116. Silent cerebral infarcts (SCI) documented by radiologic changes have been reported in 10% to 30% of patients with SCD116 and a prevalence of SCI of 53% has been documented in adults with SCD117. The prevalence of clinical stroke in SCD is reported to be as high as 24% and a crippling CVOE can occur as early as two years of age, with the highest incidence being within the first decade of life118,114. The incidence of stroke has been demonstrated to be 10% per 36 months period in children with SCD who have abnormal transcranial Doppler ultrasound (TCD)119.

The current standard of care aimed at limiting stroke episodes includes annual TCD evaluations for all children with SCD. Those children showing elevated blood flow velocities indicative of cerebrovascular narrowing and children who have experienced a stroke are treated by regular (approximately monthly) transfusion of RBCs114,116. RBC transfusions have been shown to substantially reduce the risk of stroke in SCD patients, but are associated with significant complications such as iron overload and alloimmunization. No other therapy, including hydroxyurea, has been demonstrated to be effective in the primary prevention of stroke in SCD patients, although in SCD patients previously impacted by a stroke, hydroxyurea combined with phlebotomy, when compared under clinical trial with transfusions and chelation therapy, showed non-inferiority120.

4.1. The potential for HO-1 and CO to limit Cerebro-vascular Occlusive Episodes in Sickle Cell Disease

Neuroprotective strategies that aim to reduce neuronal damage are an important approach to improving CVOE outcomes in SCD. These strategies have been explored and primarily carried out in non-sickle cell animal models including stroke, autoimmune inflammation, and traumatic and non-traumatic brain injury. Traditional neuroprotective strategies target the acute ischemic event in hopes of lessening the severity of brain injury. More recent data suggest that post-ischemic inflammation121 also contributes to brain injury following stroke, providing a possible target for neuroprotective agents, even when delivered in a delayed time window. The potential involvement of the HO/CO pathway in CVOE may include both a prevention and treatment modality. First, in preventing VOCs across the vasculature in all organs, as is discussed above, CVOEs will also likely be prevented. Second, HO and CO have been shown to mitigate the sequelae of CVOEs that occur in a number of non-SCD animal models of neural injury, that to date have included models of cerebral infarction and cerebral inflammation, among others.

4.2. HO and CO in Cerebral Ischemia

Data from six studies from four independent laboratories69,122,123,,124,125,126 implicate HO-1 and CO in limiting non-SCD cerebral ischemia and suggest that targeting this pathway will offer a novel approach to the treatment of CVOE in SCD. Additional research provides support for these observations.

Panahian and colleagues first showed that HO-1 provides neuroprotection in cerebral ischemia123. Transgenic mice overexpressing HO-1 in neurons had smaller infarcts and less cerebral edema compared to wild type mice; HO-1 was shown to impart anti-apoptotic and anti-oxidant actions in the brain mediated by inhibition of p53 protein nuclear localization and accompanied by decreases in iron staining and tissue lipid peroxidation. HO-1 was also necessary to protect the brain following N-methyl-D-aspartate (NMDA) injection, as Hmox1−/− mice had significantly greater lesion volume than wild-type littermate controls. In vitro data corroborated these results: survival after NMDA exposure was reduced in Hmox1−/− mouse neurons compared to wild-type neurons. This research substantiated a study demonstrating that overexpression of HO-1 using an adenoviral vector, attenuated brain damage after focal cerebral ischemia in rats125.

A number of studies have extended this initial work on HO-1 by investigating the effects of exogenous CO administration. In a model of transient focal cerebral ischemia, inhaled CO at the time of reperfusion reduced infarct size at 48 hours to a similar degree as that observed in transgenic mice overexpressing HO-169. Moreover, CO treatment was associated with improved neurologic deficit scores at 24 and 48 hours. This observation was supported using an intravenous, PEGylated COHb in a transient focal cerebral ischemia model. COHb levels of 11% were associated with reduced infarct volume and better neurologic deficit scores at 24 hours compared to animals that received either no transfusion or transfusion with PEG-albumin control122. Wang and colleagues124 extended the work by Zeynalov using a cerebral vascular occlusion model and inhaled CO in wild-type and Nrf2−/− mice. Administration of 125 and 250 ppm inhaled CO to wild-type mice at the onset of reperfusion and extended over 20 hours significantly reduced cerebral infarct size and resulted in better behavioral scores at 7 days post-ischemia compared to untreated mice. CO administration resulted in Nrf2 dissociation from Keap1, Nrf2 nuclear translocation, increased binding of Nrf2 to Hmox1 DNA antioxidant response elements, and increased HO-1 expression. The neuroprotective effects of CO were completely abolished in Nrf2−/− mice. Zhang evaluated the effects of CO in a 2-hour middle cerebral artery occlusion (MCAO) model in rats using CO delivered via two different types of cell free Hb126. The study demonstrated decreased infarct size after COHb administration (with approximately 5% peak COHb levels) 3 days after MCAO as well as increased blood flow to the infarct border zone. Importantly, these studies demonstrate that administration of CO, using various delivery strategies, has similar and significant neuroprotective effects after acute ischemic stroke.

4.3. HO and CO in Cerebral Inflammation

Abundant data indicate that low CO levels modulate the immune response to limit detrimental autoimmune neuroinflammation. Following ischemic stroke there is a breakdown in the blood-brain barrier that allows cells of the systemic immune system to gain access to the ischemic brain125,127,128. Antigens that are normally sequestered from the immune system may thus be encountered in the brain as well as in peripheral lymphoid organs. Indeed, by 24 hours after the induction of experimental cerebral ischemia, lymphocytes are found within the central nervous system (CNS) and CNS antigens (for example proteolipid protein) are being presented to the immune system in cervical lymph nodes129. These observations suggest that an autoimmune response to brain antigens could develop after stroke. In fact, studies in the 1970’s showed that stroke survivors had more robust cellular immune responses towards myelin basic protein (MBP) and “brain antigens” than patients with multiple sclerosis and acute inflammatory demyelinating polyneuropathy130,131,132. More recent data demonstrate lymphocyte responses to MBP and antibody responses to neurofilament and portions of the N-methyl-D-aspartate receptor among stroke survivors133,134,135. These immune responses have been largely considered an epiphenomenon of cerebral tissue injury.

In experimental and clinical studies both animals and patients who develop Th1 immune responses in the brain, suffer worse stroke outcomes121,136,137,138. It has also been shown that Th17 responses negatively impact stroke outcome139,140,141,142,143,144. These data suggest that the post-ischemic immune responses affect outcome. It has been shown that induction of MBP-specific Treg responses prior to ischemia prevents Th1 responses to MBP and improves outcome137,145,146. Therapies that can be initiated in a delayed time frame after stroke onset, especially when tissue plasminogen activator is not indicated, including CVOE in SCD, are sorely needed and the modulation of the immune response is an intervention that has potential for improving outcome.

In addition to the potential neuroprotective properties of CO, there are abundant data suggesting that CO modulates immune responses and limits detrimental autoimmune inflammation. CO has multiple effects on the immune system, which in sum, are associated with decreases in inflammation and detrimental Th1 and Th17 responses. Importantly, as pertains to the systemic encounter of CNS antigens after stroke, CO inhibits the capacity of dendritic cells to present soluble antigens to T cells147, which may, in part, be related to a block in the up-regulation of TLR4148. CORMs increased expression of HO-1, reducing the secretion of IFN-γ and IL-17 from T cells149. CO also blocks the production of IL-2 in naïve T cells150, thereby inhibiting T cell proliferation. These effects are well demonstrated in models of experimental allergic encephalomyelitis (EAE). In the EAE model, these effects were associated with a decrease in CD8 T cell accumulation in the CNS, a decrease in MHCII expression in the CNS, inhibition of Th cell proliferation and effector cell function, and a decrease in myelin-reactive T cell proliferation146. Of note, the effects of CO appear to be directed towards inhibition of Th1 and Th17 responses as opposed to up-regulation of Treg responses as the secretion of Treg cytokines was not affected149. Given the inhibitory effects of CO on the development of the Th1 and Th17 immune responses, and the fact that it can halt ongoing EAE, CO appears poised to be an effective immune modulator in post-ischemic settings including SCD-associated CVOE.

4.4. HO and CO in Brain Injury

A number of studies in various in vitro and in vivo models of cerebral injury demonstrate that CO provides neuroprotection, providing support for the use of CO in the treatment of SCD-associated CVOE’s. Four studies focused on in vitro cellular injury models in neurons, astrocytes and microglia demonstrated that CO prevents apoptosis151,152,153,154. Vieira and colleagues demonstrated that preconditioning with CO gas inhibits apoptosis in murine cerebellar granule cells treated with either glutamate or tert-butylhydroperoxide (t-BHP) to mimic excitotoxicity and oxidative stress, respectively. CO treatment induced de novo protein synthesis in these cells. This study also demonstrated that CO is a critical component of the anti-apoptotic role of HO-1, as it was found that exogenous CO administration could reverse the lack of HO-1 in an oxidative stress model. Queiroga et al, found that the anti-apoptotic properties of CO extended to astrocytes where it prevented apoptosis induced by either t-BHP or diamide, a thiol crosslinking agent. CO inhibited increases in mitochondrial membrane permeability, a key event in the intrinsic apoptotic pathway. Almeida and colleagues confirmed these effects showing that CO stimulates oxidative phosphorylation and improves metabolism, in part by preventing caspase-3 activation, mitochondrial depolarization, and changes in plasma membrane permeability 153. Additionally, Bani-Hani and colleagues showed that CO treatment using a CORM attenuated thrombin-induced activation of BV-2 microglia and reduced lactate dehydrogenase, a marker for cell breakdown154. Moreover, inhibition of HO-1 did not change the activity of CO, suggesting that CO is directly responsible for the observed neuroprotective effects. Finally, Yabluchansky et al utilized CORM-3 in a rat model of hemorrhagic stroke to demonstrate that CO reduces brain damage via a modification of the inflammatory response155.

Three in vivo studies in models of cerebral injury have been described156,157,158. The first report demonstrated the cerebro-protective effects of CO, delivered using a CORM, in a seizure-induced neonatal vascular injury model in piglets156. In this study, CO pretreatment, in contrast to air-treated control animals, prevented the loss of postictal cerebrovascular reactivity (indicative of cerebrovascular injury) to selective physiologically relevant vasodilators, both endothelium-dependent and endothelium-independent. Moreover, CO was shown to elicit vasodilator properties on the cerebral circulation, protecting the brain from cerebrovascular injury caused by seizures. The second study extended this finding, demonstrating that inhaled CO provided cerebral cytoprotection in a model of deep hypothermic circulatory arrest in piglets. CO pre-treatment completely abrogated cell death in the neocortex and hippocampus compared to air-treated control animals. CO-treated animals, in contrast to air-treated controls, maintained normal oxygen/glucose and lactate/glucose indices and corresponding cerebral sinus pressures with no change in systemic hemodynamics or metabolic intermediates suggesting that CO was acting, in part, to preserve cellular bioenergetics157. The third study demonstrated that CO, whether inhaled or delivered as a CORM, reduced cell death and ameliorated the progression of neurological deficits in a mouse model of traumatic brain injury (TBI). Treatment with CO after TBI significantly reduced pericyte death and improved functional recovery as compared to vehicle-treated control animals. In addition, CO-treated animals showed higher levels of phosphorylated neural nitric oxide synthase within neural stem cells. This study suggests that CO treatment prevents pericyte death and promotes neurogenesis158. Collectively, these data suggest that chronic administration of CO, in addition to preventing VOCs, could also mitigate the morbidities caused by specific organ ischemia, including the brain in SCD patients.

Despite the accumulating and abundant encouraging preclinical data that support the potential impact of HO and CO on improving SCD-CVOE outcomes, studies in human subjects with SCD need to proceed with caution given the host of SCD-associated organ morbidities. For example, cerebral blood flow reserve in SCD patients is often reduced and, given the possible reduction in cerebral oxygen reserve that may occur under raised CO-Hb levels, carefully constructed adequately controlled studies will be required in order to determine the risk/benefit ratio with CO in these patients.

5.0. Methods of CO Administration

Although there is substantial support for the potential therapeutic use of CO for the treatment and prevention of VOCs in SCD, going back as far as 196375, this molecule has not yet advanced to licensure as a therapeutic agent for SCD. One reason for this is the difficulty in administering CO in a chronic, environmentally safe, appropriately dosed and controlled manner. Three approaches to CO administration have been tested, including inhaled CO, CORMs, and an oral formulation of CO. Both CORMs and an oral formulation of CO are currently in development for the treatment and prevention of VOCs, respectively.

5.1. Inhaled CO

Although inhaled CO is the most obvious form of CO administration, this presents substantial challenges for further development, especially for chronic use that would be required for the prevention of VOCs. CO gas for inhalation is readily and commercially available and this route of administration has been used to generate the vast majority of preclinical and clinical data regarding CO and SCD. Inhaled CO, however, is difficult to accurately dose given the variability of patient ventilation, mask and cannula compliance, and pulmonary absorption associated with the variable capacity of the lung. Inhaled CO also has environmental safety concerns, as it requires the presence of large amounts of CO gas in cylinders. In addition, the chronic use of CO gas in an outpatient or home environment, as would be necessary for the use in the prevention of SCD VOCs, has significant logistical and compliance hurdles for patients, and potential exposure risks to others in close proximity. Despite the above challenges, the administration of inhaled CO is being utilized in a number of ongoing non-SCD targeted Phase 1 and Phase 2 clinical studies, primarily in acute indications (www.clinicaltrials.gov).

5.2. CO Releasing Molecules (CORMs)

CORMs, also termed CO prodrugs, where CO is bound to carrier molecules, including both small carrier molecules and large carrier molecules, have been explored as an alternative to inhaled CO. Small molecule CORMs have typically used transition metals such as molybdenum and ruthenium as well as boric acid/sodium borate as carriers for CO, first described in the pioneering work led by Motterlini and Foresti159. Although still under development, these metal-based CORMs present challenges for further development, including potential dose-limiting toxicity of the carrier molecule160,161. Based on these concerns, further small molecule CORM development has focused on encapsulated metal-based CORMs162, where the CORM is bound to a macromolecular carrier. Organic CORMs, where a small organic molecule releases CO under physiologic conditions160, and hybrid CO-RMs that are tethered to molecules that are known to induce HO-1 are also under evaluation163. These approaches as well as other, similarly innovative approaches are being explored in the development of CORMs155,163, with particular focus on minimizing toxicity and permitting dosing at levels likely to permit the evaluation of efficacy. Given the potential dose-limiting toxicity of the currently reported carrier molecules, if the CO release and stability prove to be appropriate, these small molecule CORMs would likely be targeted for the treatment of VOCs rather than for the chronic use necessary for the prevention of VOCs.

Large molecule CORMs, typically with PEGylated Hb carriers intended for intravenous administration, are being developed, and two such products (MP4CO and Sanguinate) have advanced into clinical evaluation. The Phase 1b clinical trial results of both MP4CO (no longer in development) and Sanguinate have been reported108,109 and a Phase 2 clinical trial with Sanguinate in the treatment of VOCs is ongoing (NCT02411708). However, in the case of these Hb-based compounds, the announced intent of the companies as well as the documented toxicity of cell-free Hb suggest that these molecules, should they succeed in clinical development, would be limited to acute use for treatment of VOC rather than chronic use for the prevention of VOC164. As an added issue, when heme is released from the exogenously administered hemoglobin, substantial acute changes that could result in toxicity as reported with hemoglobin solutions may occur164,165, and the potential for iron overload also exists if used for chronic treatment. Regardless, as new small and large molecule CORMs with a spectrum of chemical properties, reactivity and toxicology are being developed, these may result in novel innovative agents for this highly challenging disease.

5.3. Oral Formulation of CO

The potential to orally administer CO in a liquid formulation has been pursued to avoid the barriers to development of inhaled CO and CORMs detailed above166,167. CO is absorbed as well across gastrointestinal surfaces as across the pulmonary epithelium168. The development of a CO-containing liquid comprised of Generally Recognized as Safe (GRAS) excipients (HBI-002) was recently documented in abstract form and presented at research meetings166,169,170. The bioavailability of CO through oral administration of HBI-002 has been demonstrated in rodents, where an increase in COHb levels of up to 10% was achieved. Oral administration of CO avoids many of the issues associated with gas administration. As compared with inhaled CO, it allows more precise dosing and avoids environmental safety issues associated with compressed gas cylinders. In addition, it avoids the challenges associated with the reported CORMs, as no releasing molecule or reaction to release CO is required. Oral delivery of the liquid formulation provides a platform for feasibility and compliance given the preference for oral dosing by patients, especially for chronic administration and for the administration of a therapeutic outside of hospital settings.

6.0. Conclusions

CO is physiologically generated during the metabolism of heme by the HO enzymes and is measurable in blood, typically correlating with the stress response. There is much therapeutic potential for using CO in higher than physiological doses, but at levels below known toxicity to treat SCD patients. This has been known for over fifty years. Substantial preclinical and clinical data with CO has been generated, providing compelling support for its use as a potential therapeutic. More recent data underlying the therapeutic mechanisms of CO, including in SCD, have become available and these effects have been demonstrated in a plethora of in vitro preclinical and animal studies including multiple SCD mouse models. The data indicate that CO has key signaling impact on a host of metallo-enzymes as well as key genes that, in sum, result in modulation of inflammatory, oxidant and apoptotic effects as well as vasomotor tone and adhesive properties resulting in tissue preservation of vascular flow. However the role of CO as an anti-polymerization of HbS agent, while well-demonstrated, is not fully apparent at the low COHb levels targeted by these therapeutic approaches. Further work is needed to explore the known potential of CO to melt HbS tactoids and prevent polymerization, particularly within the pharmacokinetic dosing spectrum envisaged for this therapy and elucidating how this interacts with the signaling actions of CO. Further research is also indicated and hopefully forthcoming, to carefully elucidate the safety and benefits of this potential gasotransmitter therapy across the age spectrum of patients impacted by the host of patho-physiological complications of this devastating disease.

In summary, CO has important anti-vaso-occlusive and immunomodulatory effects that, together, provide cells and tissues with powerful protective and pro-survival systems in the setting of SCD. In particular, considerable scientific data provide evidence for a beneficial impact of CO on cerebrovascular complications, suggesting that in SCD, CO could potentially limit these highly problematic neurologic outcomes.

Acknowledgments

Funding has been received from NIH grants R43-HL131065 and R01-HL114567.

Contributor Information

Edward Gomperts, Hillhurst Biopharmaceuticals, Inc., 2029 Verdugo Blvd., #125 Montrose, CA, 91020; Phone: 818 445 5890.

John D. Belcher, University of Minnesota, 420 Delaware Street SE, MMC 480, Minneapolis, MN 55455; Phone: 612 624 2611, Fax: 612 625 6919.

Leo Otterbein, Harvard Medical School; Beth Israel Deaconess Medical Center, 3 Blackfan Circle Center for Life Sciences, #630 Boston, MA 02115; Phone: 617-735-2851.

Thomas D Coates, Children’s Hospital Los Angeles; University of Southern California, 4650 Sunset Boulevard MS #54 Los Angeles, CA 90027; Phone: 323-361-4624.

John Wood, Children’s Hospital Los Angeles; University of Southern California, 4650 Sunset Boulevard MS#34 Los Angeles, CA 90027; Phone: 323-361-5470.

Brett E. Skolnick, Hillhurst Biopharmaceuticals, Inc, 2029 Verdugo Blvd., #125 Montrose, CA, 91020, Phone: 818 445 5890.

Howard Levy, Hillhurst Biopharmaceuticals, Inc, 2029 Verdugo Blvd., #125 Montrose, CA, 91020; Phone: 818 445 5890.

Gregory M. Vercellotti, University of Minnesota, 420 Delaware Street SE, MMC 480, Minneapolis, MN 55455; Phone: 612 626 3757; Fax: 612 625 6919.

References

  • 1.Hebbel RP. Reconstructing sickle cell disease: a data-based analysis of the “hyperhemolysis paradigm” for pulmonary hypertension from the perspective of evidence-based medicine. American Journal of Hematology. 2011;86:123–154. doi: 10.1002/ajh.21952. [DOI] [PubMed] [Google Scholar]
  • 2.Cooper CE, Schaer DJ, Buehler PW, Wilson MT, Reeder BJ, Silkstone G, Svistunenko DA, Bulow L, Alayash AI. Haptoglobin binding stabilizes hemoglobin ferryl iron and the globin radical on tyrosine beta145. Antioxid Redox Signal. 2013;18:2264–2273. doi: 10.1089/ars.2012.4547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Moestrup SK, Moller HJ. CD163: a regulated hemoglobin scavenger receptor with a role in the anti-inflammatory response. Annals of Medicine. 2004;36:347–354. doi: 10.1080/07853890410033171. [DOI] [PubMed] [Google Scholar]
  • 4.Nielsen MJ, Moller HJ, Moestrup SK. Hemoglobin and heme scavenger receptors. Antioxid Redox Signal. 2010;12:261–273. doi: 10.1089/ars.2009.2792. [DOI] [PubMed] [Google Scholar]
  • 5.Muller-Eberhard U, Javid J, Liem HH, Hanstein A, Hanna M. Plasma concentrations of hemopexin, haptoglobin and heme in patients with various hemolytic diseases. Blood. 1968;32:811–815. [PubMed] [Google Scholar]
  • 6.Reiter CD, Wang X, Tanus-Santos JE, Hogg N, Cannon RO, 3rd, Schechter AN, Gladwin MT. Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease. Nature Medicine. 2002;8:1383–1389. doi: 10.1038/nm1202-799. [DOI] [PubMed] [Google Scholar]
  • 7.Morris CR, Gladwin MT, Kato GJ. Nitric oxide and arginine dysregulation: a novel pathway to pulmonary hypertension in hemolytic disorders. Curr Mol Med. 2008;8:620–632. doi: 10.2174/156652408786241447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Belcher JD, Chen C, Nguyen J, Milbauer L, Abdulla F, Alayash AI, Smith A, Nath KA, Hebbel RP, Vercellotti GM. Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease. Blood. 2014;123:377–390. doi: 10.1182/blood-2013-04-495887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bunn HF, Jandl JH. Exchange of heme among hemoglobins and between hemoglobin and albumin. Journal of Biological Chemistry. 1968;243:465–475. [PubMed] [Google Scholar]
  • 10.Umbreit J. Methemoglobin--it’s not just blue: a concise review. American Journal of Hematology. 2007;82:134–144. doi: 10.1002/ajh.20738. [DOI] [PubMed] [Google Scholar]
  • 11.Hrkal Z, Vodrazka Z, Kalousek I. Transfer of heme from ferrihemoglobin and ferrihemoglobin isolated chains to hemopexin. European Journal of Biochemistry. 1974;43:73–78. doi: 10.1111/j.1432-1033.1974.tb03386.x. [DOI] [PubMed] [Google Scholar]
  • 12.Hvidberg V, Maniecki MB, Jacobsen C, Hojrup P, Moller HJ, Moestrup SK. Identification of the receptor scavenging hemopexin-heme complexes. Blood. 2005;106:2572–2579. doi: 10.1182/blood-2005-03-1185. [DOI] [PubMed] [Google Scholar]
  • 13.Smith A, Morgan WT. Haem transport to the liver by haemopexin. Receptor-mediated uptake with recycling of the protein. Biochemical Journal. 1979;182:47–54. doi: 10.1042/bj1820047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Balla J, Jacob HS, Balla G, Nath K, Eaton JW, Vercellotti GM. Endothelial-cell heme uptake from heme proteins: induction of sensitization and desensitization to oxidant damage. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:9285–9289. doi: 10.1073/pnas.90.20.9285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Belcher JD, Marker PH, Geiger P, Girotti AW, Steinberg MH, Hebbel RP, Vercellotti GM. Low-density lipoprotein susceptibility to oxidation and cytotoxicity to endothelium in sickle cell anemia. Journal of Laboratory and Clinical Medicine. 1999;133:605–612. doi: 10.1016/s0022-2143(99)90191-9. [DOI] [PubMed] [Google Scholar]
  • 16.Camus SM, De Moraes JA, Bonnin P, Abbyad P, Le Jeune S, Lionnet F, Loufrani L, Grimaud L, Lambry JC, Charue D, Kiger L, Renard JM, Larroque C, Le Clesiau H, Tedgui A, Bruneval P, Barja-Fidalgo C, Alexandrou A, Tharaux PL, Boulanger CM, Blanc-Brude OP. Circulating cell membrane microparticles transfer heme to endothelial cells and trigger vasoocclusions in sickle cell disease. Blood. 2015;125:3805–3814. doi: 10.1182/blood-2014-07-589283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hanson MS, Piknova B, Keszler A, Diers AR, Wang X, Gladwin MT, Hillery CA, Hogg N. Methaemalbumin formation in sickle cell disease: effect on oxidative protein modification and HO-1 induction. British Journal of Haematology. 2011;154:502–511. doi: 10.1111/j.1365-2141.2011.08738.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Balla G, Jacob HS, Eaton JW, Belcher JD, Vercellotti GM. Hemin: a possible physiological mediator of low density lipoprotein oxidation and endothelial injury. Arteriosclerosis and Thrombosis. 1991;11:1700–1711. doi: 10.1161/01.atv.11.6.1700. [DOI] [PubMed] [Google Scholar]
  • 19.Dutra FF, Alves LS, Rodrigues D, Fernandez PL, de Oliveira RB, Golenbock DT, Zamboni DS, Bozza MT. Hemolysis-induced lethality involves inflammasome activation by heme. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:E4110–4118. doi: 10.1073/pnas.1405023111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Figueiredo RT, Fernandez PL, Mourao-Sa DS, Porto BN, Dutra FF, Alves LS, Oliveira MF, Oliveira PL, Graca-Souza AV, Bozza MT. Characterization of heme as activator of Toll-like receptor 4. Journal of Biological Chemistry. 2007;282:20221–20229. doi: 10.1074/jbc.M610737200. [DOI] [PubMed] [Google Scholar]
  • 21.Jeney V, Balla J, Yachie A, Varga Z, Vercellotti GM, Eaton JW, Balla G. Pro-oxidant and cytotoxic effects of circulating heme. Blood. 2002;100:879–887. doi: 10.1182/blood.v100.3.879. [DOI] [PubMed] [Google Scholar]
  • 22.Porto BN, Alves LS, Fernandez PL, Dutra TP, Figueiredo RT, Graca-Souza AV, Bozza MT. Heme induces neutrophil migration and reactive oxygen species generation through signaling pathways characteristic of chemotactic receptors. Journal of Biological Chemistry. 2007;282:24430–24436. doi: 10.1074/jbc.M703570200. [DOI] [PubMed] [Google Scholar]
  • 23.Ghosh S, Ihunnah CA, Hazra R, et al. Nonhaemopoietic Nrf2 dominantly impedes adult progression of sickle cell anemia in mice. JCI Insight. 2016;1 doi: 10.1172/jci.insight.81090. pii:epub 2016 Apr 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hidalgo A, Chang J, Jang JE, Peired AJ, Chiang EY, Frenette PS. Heterotypic interactions enabled by polarized neutrophil microdomains mediate thromboinflammatory injury. Nature Medicine. 2009;15:384–391. doi: 10.1038/nm.1939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Li J, Kim K, Hahm E, Molokie R, Hay N, Gordeuk VR, Du X, Cho J. Neutrophil AKT2 regulates heterotypic neutrophil-platelet interactions during vascular inflammation. Journal of Clinical Investigation. 2014;124:1483–1496. doi: 10.1172/JCI72305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Manwani D, Frenette PS. Vaso-occlusion in sickle cell disease: pathophysiology and novel targeted therapies. Blood. 2013;122:3892–3898. doi: 10.1182/blood-2013-05-498311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Turhan A, Weiss LA, Mohandas N, Coller BS, Frenette PS. Primary role for adherent leukocytes in sickle cell vascular occlusion: a new paradigm. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:3047–3051. doi: 10.1073/pnas.052522799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhang D, Chen G, Manwani D, Mortha A, Xu C, Faith JJ, Burk RD, Kunisaki Y, Jang JE, Scheiermann C, Merad M, Frenette PS. Neutrophil ageing is regulated by the microbiome. Nature. 2015;525:528–532. doi: 10.1038/nature15367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Osarogiagbon RU, Choong S, Belcher JD, Vercellotti GM, Paller MS, Hebbel RP. Reperfusion injury pathophysiology in sickle transgenic mice. Blood. 2000 Jul 1;96(1):314–320. [PubMed] [Google Scholar]
  • 30.Hebbel RP, Osarogiagbon R, Kaul D. The endothelial biology of sickle cell disease: inflammation and a chronic vasculopathy. Microcirculation. 2004;11:129–151. [PubMed] [Google Scholar]
  • 31.Rees DC, Williams TN, Gladwin MT. Sickle cell disease. Lancet. 2010;76:2018–2031. doi: 10.1016/S0140-6736(10)61029-X. [DOI] [PubMed] [Google Scholar]
  • 32.Embury SH, Hebbel RP, Mohandas N, Steinberg MH. Sickle Cell Disease: Basic principles and Clinical Practice. Raven Press; New York: 1994. [Google Scholar]
  • 33.Gardner K, Douiri A, Drasar E, Allman M, Mwirigi A, Awogbade M, Thein SL. Survival in adults with sickle cell disease in a high-income setting. Blood. 2016 Sep 8;128(10):1436–1438. doi: 10.1182/blood-2016-05-716910. [DOI] [PubMed] [Google Scholar]
  • 34.Platt OS, Rosenstock W, Espeland MA. Influence of sickle hemoglobinopathies on growth and development. N Eng J Med. 1984;311:7–12. doi: 10.1056/NEJM198407053110102. [DOI] [PubMed] [Google Scholar]
  • 35.Pleasants S. A moving target. Nature. 2014;515:S2. doi: 10.1038/515S2a. [DOI] [PubMed] [Google Scholar]
  • 36.National Heart, Lung, and Blood Institute. Sickle cell anemia: who is at risk? Bethesda, MD: US Department of Health and Human Services, National Institutes of Health, National Heart, Lung, and Blood Institute; 2016. Disease and conditions index. Available from: https://www.nhlbi.nih.gov/health/health-topics/topics/sca/atrisk. [Google Scholar]
  • 37.Haemoglobinopathies on the Move. Is Europe ready? Health and Migration Policy Perspectives. http://www.enerca.org and http://www.thalassaemia.org.cy.
  • 38.Piel FB, Hay SI, Gupta S, Weatherall DJ, Williams TN. Global burden of sickle cell anaemia in children under five, 2010–2050: modelling based on demographics, excess mortality, and interventions. PLoS Med. 2013;10(7):e1001484. doi: 10.1371/journal.pmed.1001484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Telen MJ. Beyond hydroxyurea: new and old drugs in the pipeline for sickle cell disease. Blood. 2016;127:810–819. doi: 10.1182/blood-2015-09-618553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Motterlini R, Otterbein LE. The therapeutic potential of carbon monoxide. Nat Rev Drug Discov. 2010;9(9):728–743. doi: 10.1038/nrd3228. [DOI] [PubMed] [Google Scholar]
  • 41.Foresti R, Bani-Hani MG, Motterlini R. Use of carbon monoxide as a therapeutic agent: promises and challenges. Intensive Care Med. 2008;34:649–658. doi: 10.1007/s00134-008-1011-1. [DOI] [PubMed] [Google Scholar]
  • 42.Balla G, Vercellotti G, Eaton JW, Jacob HS. Heme uptake by endothelium synergizes polymorphonuclear granulocyte-mediated damage. Trans Assoc Am Physicians. 1990;103:174–179. [PubMed] [Google Scholar]
  • 43.Otterbein LE, Soares MP, Yamashita K, Bach FH. Heme oxygenase-1: unleashing the protective properties of heme. Trends Immunol. 2003;24(8):449–455. doi: 10.1016/s1471-4906(03)00181-9. [DOI] [PubMed] [Google Scholar]
  • 44.Foresti R, Goatly H, Green CJ, Motterlini R. Role of heme oxygenase-1 in hypoxia-reoxygenation: requirement of substrate heme to promote cardioprotection. Am J Physiol Heart Circ Physiol. 2001;281(5):H1976–1984. doi: 10.1152/ajpheart.2001.281.5.H1976. [DOI] [PubMed] [Google Scholar]
  • 45.Abraham NG, Lin JH-C, Schwartzman ML, Levere RD, Shibahara S. The physiological significance of heme oxygenase. Int J Biochem. 1988;20:543–558. doi: 10.1016/0020-711x(88)90093-6. [DOI] [PubMed] [Google Scholar]
  • 46.Camhi SL, Alam J, Otterbein LE, Sylvester SL, Choi AMK. Induction of heme oxygenase-1 gene expression by lipopolysaccharide is mediated by AP-1 activation. Am J Resp Cell Mol Biol. 1995;13:387–398. doi: 10.1165/ajrcmb.13.4.7546768. [DOI] [PubMed] [Google Scholar]
  • 47.Maines MD. The heme oxygenase system: A regulator of second messenger gases. Annu Rev Pharmacol Toxicol. 1997;37:517–554. doi: 10.1146/annurev.pharmtox.37.1.517. [DOI] [PubMed] [Google Scholar]
  • 48.Applegate LA, Luscher P, Tyrell RM. Induction of heme oxygenase: A general response to oxidant stress in cultured mammalian cells. Cancer Res. 1991;51:974–978. [PubMed] [Google Scholar]
  • 49.Keyse SM, Tyrell RM. Heme oxygenase is the major 32-kDa stress protein induced in human skin fibroblasts by UVA radiation, hydrogen peroxide, and sodium arsenite. Proc Natl, Acad Sci USA. 1989;86:99–103. doi: 10.1073/pnas.86.1.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Lee PJ, Alam J, Sylvester SL, Imamdar N, Otterbein LE, Choi AMK. Regulation of heme oxygenase-1 expression in vivo and in vitro in hyperoxic lung injury. Am J Respir Cell Mol Biol. 1996;14:556–468. doi: 10.1165/ajrcmb.14.6.8652184. [DOI] [PubMed] [Google Scholar]
  • 51.Belcher JD, Young M, Chen C, Nguyen J, Burhop K, Tran P, Vercellotti GM. MP4CO, a pegylated hemoglobin saturated with carbon monoxide, is a modulator of HO-1, inflammation and vaso-occlusion in transgenic sickle cell mice. Blood. 2013;122:2757–2764. doi: 10.1182/blood-2013-02-486282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Renkens JJ, Rouflart MM, Christiaans MH, van den Berg-Loonen EM, van Hooff JP, van Heurn LW. Outcome of nonheart-beating donor kidneys with prolonged delayed graft function after transplantation. Am J Transplant. 2005;5(11):2704–2709. doi: 10.1111/j.1600-6143.2005.01072.x. [DOI] [PubMed] [Google Scholar]
  • 53.Tenhunen R, Marver HS, Schmidt R. The enzymatic catabolism of hemoglobin: stimulation of microsomal heme oxygenase by hemin. J Lab Clin Med. 1970;75:410–421. [PubMed] [Google Scholar]
  • 54.Otterbein LE, Bach FH, Alam J, et al. Carbon monoxide has anti-inflammatory effects involving the mitogen-activated protein kinase pathway. Nature Med. 2000;4:422–428. doi: 10.1038/74680. [DOI] [PubMed] [Google Scholar]
  • 55.Kim KM, Pae HO, Zheng M, Park R, Kim YM, Chung HT. Carbon monoxide induces heme oxygenase-1 via activation of protein kinase R-like endoplasmic reticulum kinase and inhibits endothelial cell apoptosis triggered by endoplasmic reticulum stress. Circ Res. 2007 Oct 26;101(9):919–927. doi: 10.1161/CIRCRESAHA.107.154781. [DOI] [PubMed] [Google Scholar]
  • 56.Chi PL, Lin CC, Chen YW, Hsiao LD, Yang CM. CO Induces Nrf2-Dependent Heme Oxygenase-1 Transcription by Cooperating with Sp1 and c-Jun in Rat Brain Astrocytes. Mol Neurobiol. 2015;52(1):277–292. doi: 10.1007/s12035-014-8869-4. [DOI] [PubMed] [Google Scholar]
  • 57.Shin DY, Chung J, Joe Y, Pae HO, Chang KC, Cho GJ, Ryter SW, Chung HT. Pretreatment with CO-releasing molecules suppresses hepcidin expression during inflammation and endoplasmic reticulum stress through inhibition of the STAT3 and CREBH pathways. Blood. 2012 Mar 15;119(11):2523–2532. doi: 10.1182/blood-2011-07-366690. [DOI] [PubMed] [Google Scholar]
  • 58.Morse D, Lin L, Choi AM, Ryter SW. Heme oxygenase-1, a critical arbitrator of cell death pathways in lung injury and disease. Free Radic Biol Med. 2009 Jul 1;47(1):1–12. doi: 10.1016/j.freeradbiomed.2009.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Chung J, Shin DY, Zheng M, Joe Y, Pae HO, Ryter SW, Chung HT. Carbon monoxide, a reaction product of heme oxygenase-1, suppresses the expression of C-reactive protein by endoplasmic reticulum stress through modulation of the unfolded protein response. Mol Immunol. 2011 Sep;48(15–16):1793–1799. doi: 10.1016/j.molimm.2011.05.014. [DOI] [PubMed] [Google Scholar]
  • 60.Sawle P, Foresti R, Mann BE, Johnson TR, Green CJ, Motterlini R. Carbon monoxide-releasing molecules (CO-RMs) attenuate the inflammatory response elicited by lipopolysaccharide in RAW264.7 murine macrophages. Br J Pharmacol. 2005 Jul;145(6):800–810. doi: 10.1038/sj.bjp.0706241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zhang X, Shan P, Alam J, Fu XY, Lee PJ. Carbon monoxide differentially modulates STAT1 and STAT3 and inhibits apoptosis via a phosphatidylinositol 3-kinase/Akt and p38 kinase-dependent STAT3 pathway during anoxia-reoxygenation injury. J Biol Chem. 2005 Mar 11;280(10):8714–8721. doi: 10.1074/jbc.M408092200. [DOI] [PubMed] [Google Scholar]
  • 62.Stewart RD. The effect of carbon monoxide on humans. Annu Rev Pharmacol. 1975;15:409–423. doi: 10.1146/annurev.pa.15.040175.002205. [DOI] [PubMed] [Google Scholar]
  • 63.Wilbur S, Williams M, Williams R, et al. Toxicological Profile for Carbon Monoxide. Atlanta (GA): Agency for Toxic Substances and Disease Registry (US); Jun, 2012. [PubMed] [Google Scholar]
  • 64.World Health Organization. Environmental Health Criteria 213 Carbon Monoxide. 2. Geneva: 1999. [Google Scholar]
  • 65.Levitt DG, Levitt MD. Carbon monoxide: a critical quantitative analysis and review of the extent and limitations of its second messenger function. Clin Pharmacol. 2015;7:37–56. doi: 10.2147/CPAA.S79626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Prabhakar NR, Dinerman JL, Agani FH, Snyder SH. Carbon monoxide: A role in carotid body chemoreception. Proc Natl Acad Sci USA. 1995;92:1994–1997. doi: 10.1073/pnas.92.6.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Sato K, Balla J, Otterbein L, Smith RN, Brouard S, Lin Y, Csizmadia E, Sevigny J, Robson SC, Vercellotti G, Choi AM, Bach FH, Soares MP. Carbon monoxide generated by heme oxygenase-1 suppresses the rejection of mouse-to-rat cardiac transplants. J Immunol. 2001;166:4185–4194. doi: 10.4049/jimmunol.166.6.4185. [DOI] [PubMed] [Google Scholar]
  • 68.Otterbein LE, Mantell LL, Choi AM. Carbon monoxide provides protection against hyperoxic lung injury. Am J Physiol. 1999;276:L688–L694. doi: 10.1152/ajplung.1999.276.4.L688. [DOI] [PubMed] [Google Scholar]
  • 69.Zeynalov E, Dore S. Low doses of carbon monoxide protect against experimental focal brain ischemia. Neurotoxicity research. 2009;15:133–137. doi: 10.1007/s12640-009-9014-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Kuramitsu K, Gallo D, Yoon M, Chin BY, Csizmadia E, Hanto DW, Otterbein LE. Carbon monoxide enhances early liver regeneration in mice after hepatectomy. Hepatology. 2011;53:2016–2026. doi: 10.1002/hep.24317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Schallner N, Pandit R, LeBlanc R, 3rd, Thomas AJ, Ogilvy CS, Zuckerbraun BS, Gallo D, Hanafy KA, Otterbein LE. Microglia regulate blood clearance in subarachnoid hemorrhage by heme oxygenase-1. J Clin Invest. 2015;125:2609–2615. doi: 10.1172/JCI78443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Fujita T, Toda K, Karimova A, Yan SF, Naka Y, Yet SF, Pinsky DJ. Paradoxical rescue from ischemic lung injury by inhaled carbon monoxide driven by derepression of fibrinolysis. Nat Med. 2001;7:598–604. doi: 10.1038/87929. [DOI] [PubMed] [Google Scholar]
  • 73.Aroutiounian SK, Louderback JG, Ballas SK, Kim-Shapiro DB. Evidence for carbon monoxide binding to sickle cell polymers during melting. Biophys Chem. 2001;91(2):167–181. doi: 10.1016/s0301-4622(01)00168-5. [DOI] [PubMed] [Google Scholar]
  • 74.Chung LL, Magdorff-Fairchild B. Extent of polymerization in partially liganded sickle hemoglobin. Archives of Biochemistry and Biophysics. 1978;189(2):535–539. doi: 10.1016/0003-9861(78)90244-8. [DOI] [PubMed] [Google Scholar]
  • 75.Sirs JA. The use of carbon monoxide to prevent sickle-cell formation. Lancet. 1963;1:971–972. doi: 10.1016/s0140-6736(63)91751-3. [DOI] [PubMed] [Google Scholar]
  • 76.Higgins JM, Eddington DT, Bhatia SN, Mahadevan L. Sickle cell vaso-occlusion and rescue in a microfluidic device. Proc Natl Acad Sci USA. 2007;104:20496–20500. doi: 10.1073/pnas.0707122105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Vandorpe DH1, Xu C, Shmukler BE, Otterbein LE, Trudel M, Sachs F, Gottlieb PA, Brugnara C, Alper SL. Hypoxia activates a Ca2+-permeable cation conductance channel sensitive to CO and to GsMTx-4 in human and mouse sickle erythrocytes. PLoS One. 2010 Jan 15;5(1):e8732. doi: 10.1371/journal.pone.0008732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Meng F, Alayash AI. Determination of extinction coefficients of human hemoglobin in various redox states. Anal Biochem. 2017 Mar 15;521:11–19. doi: 10.1016/j.ab.2017.01.002. Epub 2017 Jan 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kassa T, Jana S, Meng F, Alayash AI. Differential heme release from various hemoglobin redox states and the upregulation of cellular heme oxygenase-1. FEBS Open Bio. 2016 Aug 8;6(9):876–884. doi: 10.1002/2211-5463.12103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Nakahira K, Kim HP, Geng XH, Nakao A, Wang X, Murase N, Drain PF, Wang X, Sasidhar M, Nabel EG, Takahashi T, Lukacs NW, Ryter SW, Morita K, Choi AM. Carbon monoxide differentially inhibits TLR signaling pathways by regulating ROS-induced trafficking of TLRs to lipid rafts. J Exp Med. 2006 Oct 2;203(10):2377–2389. doi: 10.1084/jem.20060845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Chin BY, Jiang G, Wegiel B, Wang HJ, Macdonald T, Zhang XC, Gallo D, Cszimadia E, Bach FH, Lee PJ, Otterbein LE. Hypoxia-inducible factor 1alpha stabilization by carbon monoxide results in cytoprotective preconditioning. Proc Natl Acad Sci U S A. 2007 Mar 20;104(12):5109–5114. doi: 10.1073/pnas.0609611104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Bilban M, Bach FH, Otterbein SL, Ifedigbo E, d’Avila JC, Esterbauer H, Chin BY, Usheva A, Robson SC, Wagner O, Otterbein LE. Carbon monoxide orchestrates a protective response through PPARgamma. Immunity. 2006 May;24(5):601–610. doi: 10.1016/j.immuni.2006.03.012. [DOI] [PubMed] [Google Scholar]
  • 83.Bilban M, Haschemi A, Wegiel B, Chin BY, Wagner O, Otterbein LE. Heme oxygenase and carbon monoxide initiate homeostatic signaling. J Mol Med. 2008 Mar;86(3):267–279. doi: 10.1007/s00109-007-0276-0. [DOI] [PubMed] [Google Scholar]
  • 84.Haschemi A, Chin BY, Jeitler M, Esterbauer H, Wagner O, Bilban M, Otterbein LE. Carbon monoxide induced PPARγ SUMOylation and UCP2 block inflammatory gene expression in macrophages. PLoS One. 2011;6(10):e26376. doi: 10.1371/journal.pone.0026376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ferreira A, Marguti I, Bechmann I, Jeney V, Chora A, Palha NR, Rebelo S, Henri A, Beuzard Y, Soares MP. Sickle hemoglobin confers tolerance to Plasmodium infection. Cell. 2011;145:398–409. doi: 10.1016/j.cell.2011.03.049. [DOI] [PubMed] [Google Scholar]
  • 86.Wang XM, Kim HP, Nakahira K, Ryter SW, Choi AM. The heme oxygenase-1/carbon monoxide pathway suppresses TLR4 signaling by regulating the interaction of TLR4 with caveolin-1. J Immunol. 2009 Mar 15;182(6):3809–3818. doi: 10.4049/jimmunol.0712437. [DOI] [PubMed] [Google Scholar]
  • 87.Rocuts F, Ma Y, Zhang X, Gao W, Yue Y, Vartanian T, Wang H. Carbon monoxide suppresses membrane expression of TLR4 via myeloid differentiation factor-2 in betaTC3 cells. J Immunol. 2010 Aug 15;185(4):2134–2139. doi: 10.4049/jimmunol.0902782. [DOI] [PubMed] [Google Scholar]
  • 88.Riquelme SA1, Bueno SM, Kalergis AM. Carbon monoxide down-modulates Toll-like receptor 4/MD2 expression on innate immune cells and reduces endotoxic shock susceptibility. Immunology. 2015 Feb;144(2):321–332. doi: 10.1111/imm.12375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Jung SS, Moon JS, Xu JF, Ifedigbo E, Ryter SW, Choi AM, Nakahira K. Carbon monoxide negatively regulates NLRP3 inflammasome activation in macrophages. Am J Physiol Lung Cell Mol Physiol. 2015 May 15;308(10):L1058–1067. doi: 10.1152/ajplung.00400.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Wang P, Huang J, Li Y, Chang R, Wu H, Lin J, Huang Z. Exogenous Carbon Monoxide Decreases Sepsis-Induced Acute Kidney Injury and Inhibits NLRP3 Inflammasome Activation in Rats. Int J Mol Sci. 2015 Aug 31;16(9):20595–20608. doi: 10.3390/ijms160920595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Jiang L, Fei D, Gong R, Yang W, Yu W, Pan S, Zhao M, Zhao M. CORM-2 inhibits TXNIP/NLRP3 inflammasome pathway in LPS-induced acute lung injury. Inflamm Res. 2016 Nov;65(11):905–915. doi: 10.1007/s00011-016-0973-7. [DOI] [PubMed] [Google Scholar]
  • 92.Zhang W, Tao A, Lan T, Cepinskas G, Kao R, Martin CM, Rui T. Carbon monoxide releasing molecule-3 improves myocardial function in mice with sepsis by inhibiting NLRP3 inflammasome activation in cardiac fibroblasts. Basic Res Cardiol. 2017 Mar;112(2):16. doi: 10.1007/s00395-017-0603-8. doi:10.1007. [DOI] [PubMed] [Google Scholar]
  • 93.Brüne B, Ullrich V. Inhibition of platelet aggregation by carbon monoxide is mediated by activation of guanylate cyclase. Mol Pharmacol. 1987;32(4):497–504. [PubMed] [Google Scholar]
  • 94.Stone JR, Marletta MA. Soluble guanylate cyclase from bovine lung: activation with nitric oxide and carbon monoxide and spectral characterization of the ferrous and ferric states. Biochemistry. 1994 May 10;33(18):5636–5640. doi: 10.1021/bi00184a036. [DOI] [PubMed] [Google Scholar]
  • 95.Wegiel B, Gallo DJ, Raman KG, Karlsson JM, Ozanich B, Chin BY, Tzeng E, Ahmad S, Ahmed A, Baty CJ, Otterbein LE. Nitric oxide-dependent bone marrow progenitor mobilization by carbon monoxide enhances endothelial repair after vascular injury. Circulation. 2010 Feb 2;121(4):537–548. doi: 10.1161/CIRCULATIONAHA.109.887695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zuckerbraun BS, Chin BY, Wegiel B, Billiar TR, Czsimadia E, Rao J, Shimoda L, Ifedigbo E, Kanno S, Otterbein LE. Carbon monoxide reverses established pulmonary hypertension. J Exp Med. 2006 Sep 4;203(9):2109–2119. doi: 10.1084/jem.20052267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Liu XM, Chapman GB, Peyton KJ, Schafer AI, Durante W. Carbon monoxide inhibits apoptosis in vascular smooth muscle cells. Cardiovasc Res. 2002 Aug 1;55(2):396–405. doi: 10.1016/s0008-6363(02)00410-8. [DOI] [PubMed] [Google Scholar]
  • 98.Li MH, Cha YN, Surh YJ. Carbon monoxide protects PC12 cells from peroxynitrite-induced apoptotic death by preventing the depolarization of mitochondrial transmembrane potential. Biochem Biophys Res Commun. 2006 Apr 14;342(3):984–990. doi: 10.1016/j.bbrc.2006.02.046. [DOI] [PubMed] [Google Scholar]
  • 99.Kim HS, Loughran PA, Kim PK, Billiar TR, Zuckerbraun BS. Carbon monoxide protects hepatocytes from TNF-alpha/Actinomycin D by inhibition of the caspase-8-mediated apoptotic pathway. Biochem Biophys Res Commun. 2006 Jun 16;344(4):1172–1178. doi: 10.1016/j.bbrc.2006.03.180. [DOI] [PubMed] [Google Scholar]
  • 100.Wang X, Wang Y, Kim HP, Nakahira K, Ryter SW, Choi AM. Carbon monoxide protects against hyperoxia-induced endothelial cell apoptosis by inhibiting reactive oxygen species formation. J Biol Chem. 2007 Jan 19;282(3):1718–1726. doi: 10.1074/jbc.M607610200. [DOI] [PubMed] [Google Scholar]
  • 101.Beckman J, Belcher JD, Vineyard JV, Chen C, Nguyen J, Nwaneri MD, O’Sullivan MG, Gulbahee E, Hebbel RP, Vercellotti GM. Inhaled carbon monoxide reduces leukocytosis in a murine model of sickle cell disease. Am J Physiol Heart Circ Physiol. 2009;297:H1243–H1253. doi: 10.1152/ajpheart.00327.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Bains SK, Foresti R, Howard J, Atwal S, Green CJ, Motterlini R. Human sickle cell blood modulates endothelial heme oxygenase activity: effects on vascular adhesion and reactivity. Arterioscler Thromb Vasc Biol. 2010 Feb;30(2):305–312. doi: 10.1161/ATVBAHA.109.196360. [DOI] [PubMed] [Google Scholar]
  • 103.Belcher JD, Mahaseth H, Welch TE, Otterbein LE, Hebbel RP, Vercellotti GMJ. Heme oxygenase-1 is a modulator of inflammation and vaso-occlusion in transgenic sickle mice. Clin Invest. 2006;116:808–816. doi: 10.1172/JCI26857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Belcher JD, Vineyard JV, Bruzzone CM, et al. Heme-oxygenase-1 gene delivery by Sleeping Beauty inhibits vascular stasis in a murine model of sickle cell disease. J Mol Med. 2010;88:665–675. doi: 10.1007/s00109-010-0613-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Beutler E. The effect of carbon monoxide on red cell life span in sickle cell disease. Blood. 1975;46:253–259. [PubMed] [Google Scholar]
  • 106.Yallop D, Duncan ER, Norris E, Fuller GW, Thomas N, Walters J, Dick MC, Height SE, Thein SL, Rees DC. The associations between air quality and the number of hospital admissions for acute pain and sickle-cell disease in an urban environment. Br J Haematol. 2007;136(6):844–848. doi: 10.1111/j.1365-2141.2007.06493.x. [DOI] [PubMed] [Google Scholar]
  • 107.Mahan VL, Zurakowski D, Otterbein LE, Pigula FA. Inhaled carbon monoxide provides cerebral cytoprotection in pigs. PLoS One. 2012;7(8):e41982. doi: 10.1371/journal.pone.0041982. doi:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Howard J, Thein SL, Galactéros F, Inati A, Reid ME, Keipert PE, Small T, Booth F. Safety and Tolerability Of MP4CO: A Dose Escalation Study In Stable Patients With Sickle Cell Disease. American Society of Hematology. 2014 [Google Scholar]
  • 109.Misra H, Bainbridge J, Berryman J, Abuchowski A, Galvez KM, Uribe LF, Hernandez AL, Sosa NR. A phase 1b open label, randomized, safety study of SANGUINATE in patients with sickle cell anemia. Brazilian J Hematology and Hemotherapy. 2017;39:20–27. doi: 10.1016/j.bjhh.2016.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Misra H, Lickliter J, Kazo F, Abuchowski A. PEGylated carboxyhemoglobin bovine (SANGUINATE): results of a phase I clinical trial. Artif Organs. 2014 Aug;38(8):702–7. doi: 10.1111/aor.12341. [DOI] [PubMed] [Google Scholar]
  • 111.Bathoorn E, Slebos DJ, Postma DS, et al. Anti-inflammatory effects of inhaled carbon monoxide in patients with COPD: a pilot study. The European respiratory journal. 2007;30:1131–1137. doi: 10.1183/09031936.00163206. [DOI] [PubMed] [Google Scholar]
  • 112.Mayr FB, Spiel A, Leitner J, Marsik C, Germann P, Ullrich R, Wagner O, Jilma B. Effects of carbon monoxide inhalation during experimental endotoxemia in humans. Am J Respir Crit Care Med. 2005 Feb 15;171(4):354–60. doi: 10.1164/rccm.200404-446OC. Epub 2004 Nov 19. [DOI] [PubMed] [Google Scholar]
  • 113.Rhodes MA, Carraway MS, Piantadosi CA, Reynolds CM, Cherry AD, Wester TE, Natoli MJ, Massey EW, Moon RE, Suliman HB. Am J Physiol Heart Circ Physiol. Carbon monoxide, skeletal muscle oxidative stress, and mitochondrial biogenesis in humans. 2009 Jul;297(1):H392–399. doi: 10.1152/ajpheart.00164.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.DeBaun MR, Kirkham FJ. Central nervous system complications and management in sickle cell disease. Blood. 2016;127:829–838. doi: 10.1182/blood-2015-09-618579. [DOI] [PubMed] [Google Scholar]
  • 115.Schatz J, Brown RT, Pascual JM, Hsu L, Debaum MR. Poor school and cognitive functioning with silent cerebral infarcts and sickle cell disease. Neurology. 2001;56:1109–1111. doi: 10.1212/wnl.56.8.1109. [DOI] [PubMed] [Google Scholar]
  • 116.Bernaudin F, Verlhac S, Freard F, et al. Multicenter prospective study of children with sickle cell disease: radiographic and psychometric correlation. J Child Neurol. 2000;15:333–343. doi: 10.1177/088307380001500510. [DOI] [PubMed] [Google Scholar]
  • 117.Kassim AA, Pruthi S, Day M, Rodeghier M, Gindville MC, Brodsky MA, DeBaun MR, Jordan LC. Silent cerebral infarcts and cerebral aneurysms are prevalent in adults with sickle cell anemia. Blood. 2016;127:2038–2040. doi: 10.1182/blood-2016-01-694562. [DOI] [PubMed] [Google Scholar]
  • 118.Ohene-Frempong K, Weiner SJ, Sleeper LA, et al. Cerebrovascular accidents in sickle cell disease: rates and risk factors. Blood. 1998;91:288–294. [PubMed] [Google Scholar]
  • 119.Adams RJ, McKie VC, Hsu L, et al. Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial doppler ultrasonography. N Eng J Med. 1998;339:5–11. doi: 10.1056/NEJM199807023390102. [DOI] [PubMed] [Google Scholar]
  • 120.Ware RE, Helms RW, et al. Stroke with transfusion changing to hydroxyurea.(SWiTCH) Blood. 2012;118:3925–3932. doi: 10.1182/blood-2011-11-392340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Becker KJ, Kalil AJ, Tanzi P, et al. Autoimmune responses to the brain after stroke are associated with worse outcome. Stroke; a journal of cerebral circulation. 2011;42:2763–2769. doi: 10.1161/STROKEAHA.111.619593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Klaus JA, Kibler KK, Abuchowski A, Koehler RC. Early treatment of transient focal cerebral ischemia with bovine PEGylated carboxy hemoglobin transfusion. Artificial cells, blood substitutes, and immobilization biotechnology. 2010;38:223–229. doi: 10.3109/10731199.2010.488635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Panahian N, Yoshiura M, Maines MD. Overexpression of heme oxygenase-1 is neuroprotective in a model of permanent middle cerebral artery occlusion in transgenic mice. Journal of neurochemistry. 1999;72:1187–1203. doi: 10.1111/j.1471-4159.1999.721187.x. [DOI] [PubMed] [Google Scholar]
  • 124.Wang B, Cao W, Biswal S, Dore S. Carbon monoxide-activated Nrf2 pathway leads to protection against permanent focal cerebral ischemia. Stroke; a journal of cerebral circulation. 2011;42:2605–2610. doi: 10.1161/STROKEAHA.110.607101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Chao XD, Ma YH, Luo P, Cao L, Lau WB, Zhao BC, Han F, Liu W, Ning WD, Su N, Zhang L, Zhu J, Fei Z, Qu Y. Up-regulation of heme oxygenase-1 attenuates brain damage after cerebral ischemia via simultaneous inhibition of superoxide production and preservation of NO bioavailability. Experimental neurology. 2013;239:163–169. doi: 10.1016/j.expneurol.2012.09.020. [DOI] [PubMed] [Google Scholar]
  • 126.Zhang J, Cao S, Kwansa H, Crafa D, Kibler KK, Koehler RC. Transfusion of hemoglobin-based oxygen carriers in the carboxy state is beneficial during transient focal cerebral ischemia. Journal of applied physiology. 2012;113:1709–1717. doi: 10.1152/japplphysiol.01079.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Ahmad AS, Zhuang H, Dore S. Heme oxygenase-1 protects brain from acute excitotoxicity. Neuroscience. 2006;141:1703–1708. doi: 10.1016/j.neuroscience.2006.05.035. [DOI] [PubMed] [Google Scholar]
  • 128.Braun JS, Jander S, Schroeter M, Witte OW, Stoll G. Spatiotemporal relationship of apoptotic cell death to lymphomonocytic infiltration in photochemically induced focal ischemia of the rat cerebral cortex. Acta neuropathologica. 1996;92:255–263. doi: 10.1007/s004010050516. [DOI] [PubMed] [Google Scholar]
  • 129.Jander S, Kraemer M, Schroeter M, Witte OW, Stoll G. Lymphocytic infiltration and expression of intercellular adhesion molecule-1 in photochemically induced ischemia of the rat cortex. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 1995;15:42–51. doi: 10.1038/jcbfm.1995.5. [DOI] [PubMed] [Google Scholar]
  • 130.van Zwam M, Huizinga R, Melief MJ, et al. Brain antigens in functionally distinct antigen-presenting cell populations in cervical lymph nodes in MS and EAE. Journal of molecular medicine. 2009;87:273–286. doi: 10.1007/s00109-008-0421-4. [DOI] [PubMed] [Google Scholar]
  • 131.Planas AM, Gomez-Choco M, Urra X, Gorina R, Caballero M, Chamorro A. Brain-derived antigens in lymphoid tissue of patients with acute stroke. Journal of immunology. 2012;188:2156–2163. doi: 10.4049/jimmunol.1102289. [DOI] [PubMed] [Google Scholar]
  • 132.Kallen B, Nilsson O, Thelin C. Effect of encephalitogenic protein on migration in agarose of leukoytes from patients with multiple sclerosis. A longitudinal study of patients with relapsing multiple sclerosis or with cerebral infarction. Acta neurologica Scandinavica. 1977;55:47–56. [PubMed] [Google Scholar]
  • 133.Youngchaiyud U, Coates AS, Whittingham S, Mackay IR. Cellular-immune response to myelin protein: absence in multiple sclerosis and presence in cerebrovascular accidents. Australian and New Zealand journal of medicine. 1974;4:535–538. doi: 10.1111/j.1445-5994.1974.tb03233.x. [DOI] [PubMed] [Google Scholar]
  • 134.Wang WZ, Olsson T, Kostulas V, Hojeberg B, Ekre HP, Link H. Myelin antigen reactive T cells in cerebrovascular diseases. Clinical and experimental immunology. 1992;88:157–162. doi: 10.1111/j.1365-2249.1992.tb03056.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Bornstein NM, Aronovich B, Korczyn AD, Shavit S, Michaelson DM, Chapman J. Antibodies to brain antigens following stroke. Neurology. 2001;56:529–530. doi: 10.1212/wnl.56.4.529. [DOI] [PubMed] [Google Scholar]
  • 136.Dambinova SA, Khounteev GA, Izykenova GA, Zavolokov IG, Ilyukhina AY, Skoromets AA. Blood test detecting autoantibodies to N-methyl-D-aspartate neuroreceptors for evaluation of patients with transient ischemic attack and stroke. Clinical chemistry. 2003;49:1752–1762. doi: 10.1373/49.10.1752. [DOI] [PubMed] [Google Scholar]
  • 137.Kalev-Zylinska ML, Symes W, Little KC, et al. Stroke patients develop antibodies that react with components of N-methyl-D-aspartate receptor subunit 1 in proportion to lesion size. Stroke; a journal of cerebral circulation. 2013;44:2212–2219. doi: 10.1161/STROKEAHA.113.001235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Becker KJ, Kindrick DL, Lester MP, Shea C, Ye ZC. Sensitization to brain antigens after stroke is augmented by lipopolysaccharide. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2005;25:1634–1644. doi: 10.1038/sj.jcbfm.9600160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Gee JM, Kalil A, Thullbery M, Becker KJ. Induction of immunologic tolerance to myelin basic protein prevents central nervous system autoimmunity and improves outcome after stroke. Stroke; a journal of cerebral circulation. 2008;39:1575–1582. doi: 10.1161/STROKEAHA.107.501486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Gee JM, Zierath D, Hadwin J, et al. Long term immunologic consequences of experimental stroke and mucosal tolerance. Experimental & translational stroke medicine. 2009;1:3. doi: 10.1186/2040-7378-1-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Zierath D, Schulze J, Kunze A, et al. The immunologic profile of adoptively transferred lymphocytes influences stroke outcome of recipients. Journal of neuroimmunology. 2013;263:28–34. doi: 10.1016/j.jneuroim.2013.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Shichita T, Sugiyama Y, Ooboshi H, et al. Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury. Nature medicine. 2009;15:946–950. doi: 10.1038/nm.1999. [DOI] [PubMed] [Google Scholar]
  • 143.Konoeda F, Shichita T, Yoshida H, et al. Therapeutic effect of IL-12/23 and their signaling pathway blockade on brain ischemia model. Biochemical and biophysical research communications. 2010;402:500–506. doi: 10.1016/j.bbrc.2010.10.058. [DOI] [PubMed] [Google Scholar]
  • 144.Gelderblom M, Weymar A, Bernreuther C, et al. Neutralization of the IL-17 axis diminishes neutrophil invasion and protects from ischemic stroke. Blood. 2012;120:3793–3802. doi: 10.1182/blood-2012-02-412726. [DOI] [PubMed] [Google Scholar]
  • 145.Swardfager W, Winer DA, Herrmann N, Winer S, Lanctot KL. Interleukin-17 in post-stroke neurodegeneration. Neuroscience and biobehavioral reviews. 2013;37:436–447. doi: 10.1016/j.neubiorev.2013.01.021. [DOI] [PubMed] [Google Scholar]
  • 146.Hu Y, Zheng Y, Wu Y, Ni B, Shi S. Imbalance between IL-17A-producing cells and regulatory T cells during ischemic stroke. Mediators of inflammation. 2014;2014:813045. doi: 10.1155/2014/813045. doi:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Becker KJ, McCarron RM, Ruetzler C, et al. Immunologic tolerance to myelin basic protein decreases stroke size after transient focal cerebral ischemia. Proceedings of the National Academy of Sciences of the United States of America. 1997;94:10873–8. doi: 10.1073/pnas.94.20.10873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Thom SR. Carbon monoxide-mediated brain lipid peroxidation in the rat. Journal of applied physiology. 1990;68:997–1003. doi: 10.1152/jappl.1990.68.3.997. [DOI] [PubMed] [Google Scholar]
  • 149.Chora AA, Fontoura P, Cunha A, et al. Heme oxygenase-1 and carbon monoxide suppress autoimmune neuroinflammation. Journal of clinical investigation. 2007;117:438–447. doi: 10.1172/JCI28844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Tardif V, Riquelme SA, Remy S, et al. Carbon monoxide decreases endosome-lysosome fusion and inhibits soluble antigen presentation by dendritic cells to T cells. European journal of immunology. 2013;43:2832–2844. doi: 10.1002/eji.201343600. [DOI] [PubMed] [Google Scholar]
  • 151.Vieira HL, Queiroga CS, Alves PM. Preconditioning indiced by carbon monoxide provides neuronal protection against apoptosis. J neurochem. 2008;107:375–384. doi: 10.1111/j.1471-4159.2008.05610.x. [DOI] [PubMed] [Google Scholar]
  • 152.Queiroga CSF, Vercelli A, Vieira HLA. Carbon monoxide and the CNS: challenges and achievements. Brit J Pharmacol. 2015;172:1533–1545. doi: 10.1111/bph.12729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Almeida AS, Queiroga CS, Sousa MF, Alves PM, Vieira HL. Carbon monoxide modulates apoptosis by reinforcing oxidative metabolism in astrocytes: role of Bcl-2. J Biol Chem. 2012;287:10762–10770. doi: 10.1074/jbc.M111.306738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Bani-Hani MG, Greenstein D, Mann BE, Green CJ, Motterlini R. Modulation of thrombin-induced neuroinflammation in BV-2 microglia by carbon monoxide-releasing molecule 3. L Pharmacol Exp Ther. 2006;318:1315–1322. doi: 10.1124/jpet.106.104729. [DOI] [PubMed] [Google Scholar]
  • 155.Yabluchansky A, Sawle P, Homer-Vanniasinkam S, Green CJ, Foresti R, Motterlini R. CORM-3, a carbon monoxide-releasing molecule, alters the inflammatory response and reduces brain damage in a rat model of hemorrhagic stroke. Crit Care Med. 2012;40:544–552. doi: 10.1097/CCM.0b013e31822f0d64. [DOI] [PubMed] [Google Scholar]
  • 156.Zimmermann A, Leffler CW, Tcheranova D, Fedinec AL, Parfenova H. Cerebroprotective effects of the CO-releasing releasing molecule CORM-A1 against seizure-induced neonatal vascular injury. Am J Physiol Heart Circ Physiol. 2007;293:H2501–7. doi: 10.1152/ajpheart.00354.2007. [DOI] [PubMed] [Google Scholar]
  • 157.Mahan VL. Neuroprotective, neurotherapeutic and neurometabolic effects of corbon monoxide. medical gas research. 2012;2:32–36. doi: 10.1186/2045-9912-2-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Choi YK, Maki T, Mandeville ET, Koh SH, Hayakawa K, Arai K, Kim YM, Whalen MJ, Xing C, Wang X, Kim KW, Lo EH. Dual effects of carbon monoxide on pericytes and neurogenesis in traumatic brain injury. Nat Med. 2016;22(11):1335–1441. doi: 10.1038/nm.4188. [DOI] [PubMed] [Google Scholar]
  • 159.Motterlini R, Clark JE, Foresti R, Sarathchandra P, Mann BE, Green CJ. Carbon monoxide-releasing molecules: characterization of biochemical and vascular activities. Circ Res. 2002;90:E17–E24. doi: 10.1161/hh0202.104530. [DOI] [PubMed] [Google Scholar]
  • 160.Ji X, Damera K, Zheng Y, Yu B, Otterbein LE, Wang B. Toward carbon-monoxide-based therapeutics: Critical drug delivery and developmentability issues. J Pharm Sci. 2016;105:406–416. doi: 10.1016/j.xphs.2015.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Winburn IC, Gunatunga K, McKernan RD, Walker RJ, Sammut IA, Harrison JC. Cell damage following carbon monoxide releasing molecule exposure: implications for therapeutic applications. Basic & clinical pharmacology & toxicology. 2012;111:31–41. doi: 10.1111/j.1742-7843.2012.00856.x. [DOI] [PubMed] [Google Scholar]
  • 162.Inaba H, Fujita K, Ueno T. Design of biomaterials for intracellular delivery of carbon monoxide. Biomater Sci. 2015;3:1423–1438. doi: 10.1039/c5bm00210a. [DOI] [PubMed] [Google Scholar]
  • 163.Wilson JL, Kobeissi S, Oudir S, Haas B, Michel B, Dubois Rande JL, Olivier A, Martens T, Rivard M, Motterlini R, Foresti R. Design and synthesuis of new hybrid molecules that activates the transcription factor Nrf2 and simultaneously release carbon monoxide. Chemistry. 2014;20:14698–14704. doi: 10.1002/chem.201403901. [DOI] [PubMed] [Google Scholar]
  • 164.Natanson C, Kem SJ, Lurie P, Banks SM, Wolfe SM. Cell-free hemoglobin-based blood substitutes and risk of myocardial infarction and death: a meta-analysis. JAMA. 2008;299:2304–2312. doi: 10.1001/jama.299.19.jrv80007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Alayash AI. Hemoglobin-Based Blood Substitutes and the Treatment of Sickle Cell Disease: More Harm than Help? Biomolecules. 2017 Jan 4;7(1) doi: 10.3390/biom7010002. pii E2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Gomperts E, Forman HJ, Liu H, Lee M. The pharmacokinetic evaluation of oral administered carbon monoxide instilled in a liquid formulation (CO-LF) to rats to determine carbon monoxide hemoglobin levels with potential efficacy in patients with Sickle Cell Disease (SCD) American Society of Hematology. 2013 [Google Scholar]
  • 167.Otterbein LE, Foresti R, Motterlini R. Heme Oxygenase-1 and Carbon Monoxide in the Heart: The Balancing Act Between Danger Signaling and Pro-Survival. Circ Res. 2016 Jun 10;118(12):1940–1959. doi: 10.1161/CIRCRESAHA.116.306588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Coburn RF. Carbon monoxide uptake in the gut. Ann NY Academy of Sciences. 1968;150:13–21. doi: 10.1111/j.1749-6632.1968.tb19025.x. [DOI] [PubMed] [Google Scholar]
  • 169.Becker KJ, Zierath D, Shen A, Levy H, Otterbein LE, Gomperts ED, Skolnick BE. Oral Carbon Monoxide as a Treatment for Stroke. Poster session presented at the International Stroke Conference; Houston, TX. 2017. Mar, [Google Scholar]
  • 170.Belcher JD, Gomperts E, Levy H, Otterbein L, Vercellotti GM. HBI-002, an oral carbon monoxide therapeutic, is a modulator of vaso-occlusion in transgenic sickle cell mice. Foundation for Sickle Cell Disease Research. 2017 [Google Scholar]

RESOURCES