2045-9912-2-28 2045-9912 Review <p>Emerging concepts on the anti-inflammatory actions of carbon monoxide-releasing molecules (CO-RMs)</p> MotterliniRobertoroberto.motterlini@inserm.fr HaasBenjaminbenjamin.haas@inserm.fr ForestiRobertaroberta.foresti@inserm.fr

INSERM U955, Equipe 3, Faculty of Medicine, University Paris-Est Creteil, Creteil, France

Medical Gas Research 2045-9912 2012 2 1 28 http://www.medicalgasresearch.com/content/2/1/2810.1186/2045-9912-2-2823171578
4102012711201221112012 2012Motterlini et al.; licensee BioMed Central Ltd.This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Inflammation Carbon monoxide-releasing molecules (CO-RMs) Oxidative stress Inflammatory mediators Bactericidal activities

Abstract

Carbon monoxide-releasing molecules (CO-RMs) are a class of organometallo compounds capable of delivering controlled quantities of CO gas to cells and tissues thus exerting a broad spectrum of pharmacological effects. CO-RMs containing transition metal carbonyls were initially implemented to mimic the function of heme oxygenase-1 (HMOX1), a stress inducible defensive protein that degrades heme to CO and biliverdin leading to anti-oxidant and anti-inflammatory actions. Ten years after their discovery, the research on the chemistry and biological activities of CO-RMs has greatly intensified indicating that their potential use as CO delivering agents for the treatment of several pathological conditions is feasible. Although CO-RMs are a class of compounds that structurally diverge from traditional organic-like pharmaceuticals, their behaviour in the biological environments is progressively being elucidated revealing interesting features of metal-carbonyl chemistry towards cellular targets. Specifically, the presence of carbonyl groups bound to transition metals such as ruthenium, iron or manganese appears to make CO-RMs unique in their ability to transfer CO intracellularly and amplify the mechanisms of signal transduction mediated by CO. In addition to their well-established vasodilatory activities and protective effects against organ ischemic damage, CO-RMs are emerging for their striking anti-inflammatory properties which may be the result of the multiple activities of metal carbonyls in the control of redox signaling, oxidative stress and cellular respiration. Here, we review evidence on the pharmacological effects of CO-RMs in models of acute and chronic inflammation elaborating on some emerging concepts that may help to explain the chemical reactivity and mechanism(s) of action of this distinctive class of compounds in biological systems.

Introduction

The heme oxygenase enzymes (HMOX1 and HMOX2) generate, among other interesting molecules, the gas carbon monoxide (CO) 1. The quantity of the gas produced over time depends on the tissue examined, as HO-2 is constitutively expressed in the endothelium, testes and brain while HMOX1 is highly inducible in all tissues by many kinds of stressful stimuli 2. In addition, the availability of the substrate heme, which is cleaved by heme oxygenases in the α position to release CO, will strictly determine how much CO can be produced in the cell. However, it is expected that CO levels will increase upon up-regulation of HMOX1 and this has been demonstrated in some studies that directly measured CO production ex-vivo 3 4. Research conducted in the past years has progressively elucidated the role of HMOX1 in mammalian systems. The protein was recognized early on as an important component of the stress response 5 6 but it was only when detailed exploration of the function of its products was performed that an indispensable action for HMOX1 as an anti-oxidant and anti-inflammatory system emerged 7 8. CO appears to contribute most significantly to these anti-inflammatory activities by regulating a variety of transcription factors, inflammatory proteins and pathways 9 10 11. This role is consistent across many inflammatory conditions, although the specific pathways affected might differ from one disease to another.

Exposure of cells, tissues and animals to sub-toxic amounts of CO gas has been used successfully to reproduce the anti-inflammatory properties of HMOX1 and has helped to unravel many of the mechanisms underlying this effect 10 11. The use of CO gas as a therapeutic agent is also underscored by clinical trials currently ongoing in patients who will receive CO by inhalation for the treatment of pulmonary arterial hypertension, post-operative ileus and idiopathic pulmonary fibrosis (see http://www.clinicaltrials.gov). In parallel and as an alternative to this experimental approach, we have focused our strategy on utilizing chemicals that could bind and carry CO stably but deliver the gas when used in biological systems. We have identified and termed these compounds CO-releasing molecules (CO-RMs) 12 13 14 and have extensively studied their biochemical, biological and pharmacological effects in many in vitro and in vivo models of disease 9 15 16 17. The chemical structure of the best characterized CO-RMs (CORM-2, CORM-3, CORM-A1 and CORM-376) is represented in Figure  1 (see also chemical formula in List of Abbreviations). Thanks to strong collaborations with chemists, we have generated CO-RMs with improved water solubility, diverse chemical structures, various rates of CO release and stability 9 15 58 59 60. The results have been encouraging as the concentrations and doses of CO-RMs used in all studies were such that the final CO exposure was below the threshold believed to cause toxicity. In addition, as shown in Table  1, the data obtained so far support promising pharmacological actions of CO-RMs that could be useful to counteract inflammatory conditions. Metal carbonyl complexes, containing ruthenium, iron or manganese as metal center, and boranocarbonates are the two major classes of CO-RMs in our portfolio 15 16 59 61. Our own investigations and studies with collaborators revealed that metal carbonyls are better anti-inflammatory agents than boranocarbonates but the reasons for this difference are unknown at present. The chemistry and pharmacological properties of these compounds is becoming a topic of great interest since in the last 2-3 years other research groups have synthesized a variety of different new CO-RMs. Recently ruthenium imidazole oxime carbonyls 62, photoactive and nanoCO-RMs 63 64, enzyme-triggered CO-RMs 65, CO-RMs encapsulated in micelles 66 and rhenium-based CO-RMs 67 have been developed. However, for most of these new molecules a detailed picture of their behaviour in cells, tissues and in vivo models of disease is not yet available, and will be required to evaluate their full pharmacological potential. In this context it is important to emphasize that, once CO is liberated, the potential toxicity of the residual molecule containing the transition metal needs to be carefully evaluated. To date a systematic in vivo toxicological profile of CO-RMs has not yet been performed but this will be required once a lead compound will be identified for a given pathological indication.

<p>Figure 1</p>

Chemical structure of the best characterized CO-RMs which have been shown to exert anti-inflammatory and anti-bacterial activities in vitro and in vivo (see also Table  1 for more details)

Chemical structure of the best characterized CO-RMs which have been shown to exert anti-inflammatory and anti-bacterial activities in vitro and in vivo (see also Table 1 for more details).

<p>Table 1</p>

Type of CO-RM

Inflammatory disease models

Effect on inflammatory markers/

References

in vitro and in vivo

Overall outcome

CORM-3

Vascular thrombosis in rats

↓ fibrinogen and fibrin

18

↑ pro-thrombin

CORM-3

Hemorrhagic stroke induced by collagenase in rats

↓ TNF-α production

19

↓ brain injury

CORM-2

Polymicrobial sepsis induced by cecal ligation and perforation (CLP) in mice

↑ protein C system

20

↓ plasma thrombomodulin

↓ number of thrombi in liver, kidney and lung

CORM-3

Postmenopausal rheumatoid arthritis osteoporosis in mice

↓ cellular infiltration and cartilage degradation

21

↓ TNF-α production

↓ Serum levels of IL-6, alkaline phosphatase and

MMP-3

CORM-2

Coagulation and fibrinolitic markers in human umbilical vein endothelial cells (HUVEC)

↓ tissue factor

22

↓ plasminogen activator inhibitor type 1 (PAI-1)

CORM-2

Neuropatic pain and microglia activation in mice induced by nerve injury

↓ mechanical allodynia

23

CORM-3

↓ thermal hyperalgesia

↓ nNOS and iNOS expression

↓ microglial marker (CD11b/c)

CORM-3

Vascular inflammation in human umbilical vein endothelial cells (HUVEC)

↓ VCAM-1 and ECAM expression

24 25 26

CORM-2

↓ NF-kβ and p38-MAPK expression

↓ mitochondrial respiration

↓ NF-kβ and iNOS expression

CORM-2

Ischemia-reperfusion injury after kidney transplantation in rats

↑ survival rate

27

CORM-3

↓ acute tubular necrosis and hemorrhage

CORM-2

Colitis induced by dextran sodium sulfate in mice

↓ disease activity index

28

↓ myeloperoxidase (MPO) activity

↓ TNF-α production

CORM-2

Endoplasmic reticulum (ER) stress induced by thapsigargin in human hepatocytes and mice

↓ C-reactive protein (CRP)

29

CORM-3

↓ serum amyloid P component (SAP)

CORM-2

Cutaneous wound healing in rats

↑ cell proliferation and wound contraction

30

↑ collagen synthesis

↓ TNF-α production and ICAM-1 expression

↑ IL-10 production

CORM-2

Acute hepatic ischemia-reperfusion injury in rats

↑ anti-apoptotic protein Bcl2

31

↓ markers of hepatic damage (AST/ALT)

↓ serum levels of TNF-α and IL-6

↓ caspase activity and NF-kβ expression

CORM-2

Acute pancreatitis in rats

↓ serum levels of TNF-α and IL-1β

32

↓ NF-kβ expression and MPO activity

↑ IL-10 production

CORM-3

Rheumatoid arthritis induced by KBxN serum transfer in mice

↑ serum osteocalcin and HO-1 expression

33

↓ MMP-9, MMP-13 and IL-1β expression

↓ high mobility group box 1 (HMGB1)

↓ Receptor activator of nuclear factor κB ligand (RANKL)

CORM-2

Ischemia-reperfusion induced inflammation of small intestine in mice

↓ TNF-α and ICAM expression

34

↓ leukocytes rolling and adhesion

↓ NF-kβ expression and MPO activity

CORM-2

Inflammatory response induced by lipopolysaccharide in RAW 264.7 macrophages and BV-2 microglia

↓ iNOS mRNA and protein expression

35

↑ COX-2 expression and NF-kβ expression

↑ PGE2 levels

↑ phosphorylation of Akt and MAPKs

CORM-2

Sepsis induced by cecal ligation and puncture (CLP) in mice

↓ neutrophil infiltration in broncoalveolar lavage and liver

36 37 38 39

↓ MPO activity in lung and NF-kβ in liver

↓ serum TNF-α and high mobility group box 1 (HMGB1)

↓ nitrite/nitrate plasma levels

↑ cardiac PGC-1α and mitochondrial biogenesis

CORM-A1

Inflammatory response induced by TNF- α in brain vascular endothelial cells

↓ NADPH oxidase activity

40

↓ apoptosis

CORM-3

Post-operative ileus in mice

↑ intestinal contractility and transit

41

CORM-A1

↑ IL-10 and HMOX1 expression

↓ IL-6 and iNOS expression

↓ leukocyte infiltration

CORM-2

Inflammatory response in liver and small intestine of thermally injured mice

↓ TNF-α, IL-1β and iNOS expression

42 43

↓ leukocyte infiltration

↓ NF-kβ and ICAM-1 expression

CORM-3

Bacterial activity in vitro and bacterial infection in vivo

E. Coli and P. Aeuruginosa growth in vitro

17 44 45 46 47 48

CORM-2

↑ bacteriostatic and bactericidal effects in vivo

CORM-A1

↑ increased phagocytosis in vivo

CORM-371

↓ respiration in bacteria

ALF-021

↑ survival after bacterial infection in vivo

ALF-062

↓ high mobility group box 1 (HMGB1)

CORM-3

Collagen-induced arthritis in mice

↓ prostaglandin E2 (PGE2)

49

↓ pro-inflammatory interleukins (IL-1β, IL-2, IL-6)

↑ anti-inflammatory interleukins (IL-10)

↓ expression of cyclooxygenase-2 (COX-2)

CORM-3

Vascular inflammation in human neutrophils and rat primary endothelial (EC) and mast (MC) cells

↓ oxidative burst in human PMNs

50

↓ expression of CD54 in rat ECs

↓ histamine in MCs

CORM-2

Inflammatory response induced by pro-inflammatory cytokines in Caco-2 cells and human chondrocytes

↓ prostaglandin E2 (PGE2) and nitrite

51 52 53

↓ ROS production

↓ COX-2 and iNOS protein expression

↓ NF-kβ expression

↓ metalloproteinase-7 (MMP-7)

CORM-3

Leukocyte-endothelial interaction in vitro and in a models of acute pancreatitis and paw edema in mice

↓ PMNs leukocytes in peritoneal cavity

54

CORM-A1

↓ PMNs rolling on endothelial cells

↓ paw edema and swelling

CORM-3

Inflammatory response induced by LPS/INFγ and hypoxia in BV2 microglia cells

↓ nitrite and TNF-α production

55 56

↓ hypoxia-reoxygenation damage

CORM-2

Inflammatory response induced by lipopolysaccharide in RAW 264.7 macrophages

↓ nitrite and TNF-α production

57

CORM-3

Effect of CO-RMs on in vitro and in vivo inflammatory disease models

In this article we will review the role of CO-RMs in protection against inflammatory conditions, focusing primarily on their effect on oxidative stress and nitric oxide (NO) production, two of the main initiators of the inflammatory cascade. The biochemical and physiological assays used to determine CO liberation or transfer of CO to cellular targets and their interesting anti-bacterial actions will also be discussed.

Anti-inflammatory actions of CO-RMs

Inflammation and host defense are necessary and intrinsic processes that serve to protect organisms from a series of pathological challenges. The mechanisms that accompany the inflammatory response involve multiple cell types, signalling pathways and transcriptional factors and inflammation appears to be relevant for the vast majority of chronic diseases as well as in acute conditions 68. That HMOX1 is a key player in mitigating inflammation was first reported in a model of carrageenin-induced pleurisy in rats, in which the evolution of inflammation was accompanied by a dramatic increase in HMOX1 levels and inhibition of heme oxygenase activity enhanced inflammatory markers 69. In addition, HMOX1 deficiency in human subjects exhibited high levels of vascular inflammation and oxidative stress 70, a finding which is highly reproducible in mice lacking this stress protein 71. Although bilirubin and biliverdin, endowed with potent antioxidant properties, may be important contributors that fight inflammation 72 73, CO gas applied exogenously is often found to recapitulate many of the anti-inflammatory actions elicited by HMOX1 74. Our work on the discovery and characterization of CO-RMs was carried out while novel findings by Otterbein et al. described the powerful effect of CO gas in inhibiting the production of pro-inflammatory cytokines (TNF-α, IL-1β) stimulated by lypopolysaccharide (LPS) in vitro and in vivo, showing at the same time that CO induced the expression of the anti-inflammatory cytokine IL-10 and that mitogen activated protein kinases (MAPKs) mediated this phenomenon 8. This and other exciting work stimulated our efforts in the development of CO-RMs and in trying to understand their efficacy in disease models. From an anti-inflammatory perspective, CO-RMs can affect multiple cell types and pathways that coordinate the inflammatory cascade (see Table  1 for a summary of the anti-inflammatory activities of CO-RMs in various in vitro and in vivo models). For example, Urquhart et al found that CORM-3 strongly reduced neutrophil extravasation in the peritoneum of zymosan-treated mice and inhibited expression of adhesion molecules in human polymorphonuclear neutrophils (PMNs) 54. Still focusing on PMNs, Sun and co-workers showed that CORM-2 attenuated the leukocyte sequestration, Nfkβ activation and endothelial protein expression of ICAM-1 in the lung of thermally injured mice 75. The multiple effects of CO-RMs were particularly well dissected in a study by Masini et al. where human PMNs primed to elicit an inflammatory response were co-incubated with rat endothelial cells or perivascular mast cells 50. Here the authors clearly showed that CORM-3 down-regulated the oxidative burst in PMNs, the over-expression of adhesion molecules in PMNs and endothelial cells and the release of histamine and up-regulation of an activation marker by mast cells. These results indicate how CORM-3 modulates acute inflammation by reducing the activation of PMNs, the first responders in host defense, but also by inhibiting expression of molecules and inflammatory factors that perpetuate the inflammatory process. In RAW macrophages and BV-2 microglia we have also shown concentration-dependent decreases in nitrite and TNF-α production by CORM-2 and CORM-3 following challenge with LPS 55 56 57.

The in vivo anti-inflammatory action of CO-RMs has also been consistently described. The group of Alcaraz has performed a series of detailed investigations in arthritis models 21 33 49 and demonstrated that daily treatment with CORM-2 or CORM-3 can effectively suppress the clinical and histopathological manifestations of disease. Levels of PGE-2 and many other inflammatory mediators were reduced in the joint and this resulted in a better preservation of cartilage tissue and bone structures 33. However, modulation of inflammatory molecules levels surely is not the only mechanism contributing to the CO-RMs mediated protection against inflammation and the data by Lancel and colleagues point to mitochondria as very important cellular organelles that are affected by CO-RMs. In a model of sepsis induced by cecal ligation, CORM-3 administration conserved cardiac mitochondrial function by preventing sepsis-mediated damage to mitochondria thus preserving membrane potential and respiration and inducing mitochondrial biogenesis 36. In the heart of mice fed a high fat diet to mimic a metabolic syndrome-like disorder CORM-3 also stimulated mitochondrial biogenesis 76. The mode of action and efficacy of CO-RMs may also depend on the timing of administration in relation to the pathology studied, as exemplified recently by our investigation in a model of hemorrhagic stroke in rats 19. Indeed, we observed that CORM-3 pre-treatment (5 min) or post-treatment (3 days) of rats after the onset of the hemorrhage elicited protective effects while administering the compound 3 hours after the stroke, in correspondence to the acute phase of the disease process, resulted in exacerbation of damage. The striking observation of this study is that one single dose of CORM-3 could modify the long-term inflammatory scenario that followed the hemorrhagic stroke by redirecting and limiting the infiltration of peripheral leukocytes and neutrophils in the brain and reducing the local activation of brain microglia and astrocytes induced by the stroke. Importantly, CORM-3 appeared to finely tune the levels of TNF-α, by allowing its positive action in reparatory processes but inhibiting its detrimental effects. Thus, a growing body of literature supports a beneficial role of CO-RMs in inflammatory models but future investigations are required to better establish their therapeutic applications (see Figures  2 and 3 for the proposed mechanism of action of CO-RMs in vitro and in vivo).

<p>Figure 2</p>

Graphical representation summarizing the mechanism(s) potentially involved in the anti-inflammatory activities of CO-RMs (see text for details)

Graphical representation summarizing the mechanism(s) potentially involved in the anti-inflammatory activities of CO-RMs (see text for details).

<p>Figure 3</p>

Schematic diagram representing the diverse effects of CO liberated from CO-RMs in vitro and in vivo (see text for details)

Schematic diagram representing the diverse effects of CO liberated from CO-RMs in vitro and in vivo (see text for details).

Mechanisms underlying the effects of CO-RMs on inflammation: modulation by oxidative stress

It is well accepted that inflammatory stimuli promote a variety of responses that participate to exacerbate damage in cells and tissues but also promote the resolution of inflammation. Oxidative stress, derived from excessive and persistent production of reactive oxygen species (ROS) and a possible decrease in antioxidant defences, accompanies or precedes the increased amounts of inflammatory mediators upon inflammatory challenge. Because CO has a high affinity for different heme-containing proteins – cytochromes in mitochondria and NADPH oxidase in the cell 77 - that contribute to regulate the levels of ROS, it is intriguing that part of the anti-inflammatory activities of CO-RMs may derive directly from inhibiting the generation of these damaging (or signalling) species. CO-RMs have shown a tendency to modulate pathways that produce ROS and the chemical nature of transition metal carbonyls might favour this reaction by allowing a selective transfer of CO from the CO-RMs to the target 78 79. In RAW macrophages treated with LPS or PMA-stimulated neutrophils CORM-2 inhibited NADPH activity and overproduction of superoxide anion (O2) 80. Similarly, CORM-A1 reduced the accumulation of ROS induced by TNF-α in pig cerebral microvascular endothelial cells, possibly by acting on a specific subunit of NADPH oxidase (Nox4) which is highly expressed in these cells 40 81. Notably, a reduction in oxidative stress was reported also in chondrocytes from cartilage specimens of patients suffering from osteoarthritis, emphasizing both the relevance of these findings in primary human tissue and the idea that pathological processes occurring in diseased tissue can be modified by application of CO-RMs 51. Oxidative stress levels were also significantly reduced by CORM-3 in intestinal tissue in a clinically relevant model of postoperative ileus and this was accompanied by partial restoration of antioxidant capacity levels 41. Increased production of ROS following TNF-α/cycloheximide exposure was also diminished by CORM-A1 in a mouse intestinal epithelial cell line 82. In summary, different CO-RMs can inhibit ROS/oxidative stress that results from inflammation, thus affecting an early and crucial mechanism that modulates subsequent inflammatory processes (see Figure  2).

Mechanisms underlying the effects of CO-RMs on inflammation: modulation of inducible nitric oxide synthase (iNOS) and NO production

Inflammation is a complex phenomenon; thus, it is anticipated that any anti-inflammatory properties of CO-RMs would involve a number of metabolic pathway. Overproduction of NO following up-regulation of inducible NO synthase (iNOS) is a critical step in the initiation and propagation of the inflammatory response 83 and diverse actions of CO-RMs in relation to this system have been described although with quite conflicting findings. We have observed that CORM-2 and CORM-3 decrease NO levels produced by macrophages stimulated with LPS without affecting iNOS protein expression 57 and because of these results we have postulated that CO from CO-RMs inhibited the activity of iNOS, a heme-containing protein already shown in purified form to be blocked by CO gas 84. Similar results were obtained in microglia by Min KJ et al. 85 while Megias and colleagues actually demonstrated that iNOS expression was reduced by CORM-2 in Caco-2 cells challenged with a combination of IL-1β, TNF-α and IFN-γ 52. Other authors have reported the same observation in the gut 41 and in the spinal cord 23 following inflammatory damaging conditions, strengthening the idea that indeed CO-RMs exert an inhibitory effect on iNOS induction and activity. This would not perhaps be surprising if we consider that CO-RMs seem to affect the activation of Nfkβ 24 25 52, which controls the expression iNOS and is a master regulator of major pathways in inflammation. However, until detailed studies designed to dissect the effect of CO-RMs on activity versus induction of iNOS are performed, it will not be clear whether CO-RMs can inhibit NO generation or iNOS expression. It may well be that inhibition of both can occur simultaneously or that one effect or the other will prevail depending on the inflammatory condition, tissue analyzed and the type of CO-RM investigated.

Bactericidal activities of CO-RMs

The threat of bacterial infection is omnipresent in surgical settings, wounds and contaminated food, any of which may lead to fatal consequences. Interestingly, CO-RMs have been shown to possess anti-bacterial properties that may be among the important therapeutic applications envisioned for this class of compounds. Lack of HMOX1 in mice resulted in an exaggerated lethality after cecal ligation and puncture (CLP), which caused polymicrobial sepsis 86. However, the administration of CORM-2 was able to increase phagocytosis, decrease circulating bacterial counts, and rescue HMOX1–/– mice from the exaggerated mortality of CLP-induced sepsis, even when applied 6 hours after the initiation of infection. This is a remarkable result that emphasizes how these molecules can exert pleiotropic actions in such a complicated and severe pathological scenario. Desmard et al. also demonstrated that CORM-3, CORM-2 and, to a lesser extent, CORM-371, exert anti-bacterial actions against P. Aeruginosa in vitro and in vivo 17 87. The ruthenium-based CO-RMs appeared more effective and CORM-A1 only exerted a transient bacteriostatic action, highlighting again the importance of the metal in mediating some activities of CO-RMs and perhaps guiding CO to the appropriate cellular target. More detailed work has been performed to investigate the direct effect of CO-RMs on different bacteria and the results have been reviewed elsewhere 88 89.

Using more biochemical-oriented approaches, it has been possible to determine that terminal oxidases are targeted by CORM-3 when inhibiting bacterial growth 17 44, thus impairing bacterial respiration. Moreover, oxidative stress caused by CO-RMs is another factor explaining some mechanistic actions of these compounds. According to Tavares et al., exposure of E. Coli to CORM-2 or a molybdenum-containing CO-RM increased the levels of intracellular ROS as well as causing DNA damage and disruption of Fe-S clusters 45. The authors also showed that thiol-based antioxidants prevented the anti-microbial properties of CO-RMs, a finding we corroborated in studies using P. Aeruginosa 17 87. However, in our work no effect of CORM-2 or CORM-3 on ROS production was detected as assessed by the use of a fluorescent probe 17, while in another collaborative investigation it was observed that thiols reduced the ROS production stimulated by CORM-2 in P. Aeruginosa biofilms but that this reduction was not accompanied by inhibition of bacterial growth 46. Thus, the collective evidence suggests that CO-RMs interacts with metal-based proteins present in bacteria to exert various effects that are related to their bactericidal or bacteriostatic properties. However, it is possible that other pathways, susceptible to changes following application of CO-RMs, contribute to CO-RMs-mediated anti-microbial effects. In this regard, data obtained from microarray analysis of bacterial genes affected by exposing E. Coli to CORM-2 and CORM-3 have provided fascinating information about the pathways responding to CO-RMs 44 90. It should be noted that one study looked at aerobically and anaerobically grown E Coli and CORM-2 while the other investigated CORM-3 in anaerobically grown E Coli, thus already indicating that different results should be expected from this analysis. Saraiva and colleagues have nicely summarized the diverse, and perhaps still incomplete, information collected in these two transcriptomic approaches showing that some pathways are typically changed in anaerobic conditions, some only in the aerobic state and some are instead commonly altered in E Coli grown either in aerobic or anaerobic conditions 88. Of interest, genes involved in zinc homeostasis and the bacterial response to oxidative stress (SoxRS and OxyR) were increased in both conditions, perhaps stressing that, irrespective of the growth medium, the presence of ruthenium in CO-RMs and their propensity to cause oxidative stress/inhibit the respiratory complexes will consistently occur. The fact that genes modulating biofilm pathways are affected is also a clear signal that E. Coli is suffering from exposure to CO-RMs and thus attempts to boost its resistance to these agents by promoting biofilm formation. In addition, increased methionine metabolism is triggered by CO-RMs, which may still be linked to an oxidative stress response.

In summary, few, but well designed and informative reports support the idea that CO-RMs are useful compounds to be employed alone or in combination with other antibiotics 46 to fight bacterial infection, taking into account the important notion that the bactericidal actions of CO-RM-s are elicited at concentrations that do not harm mammalian cells 17.

Liberation of CO by CO-RMs: biochemical and pharmacological assays

Assessing CO liberation from CO-RMs has been a priority since our discovery of these compounds. Initially we developed a myoglobin assay for the detection of carbon monoxy myoglobin (MbCO) and employed an amperometric CO electrode to determine the rate and quantity of CO released 12 16. Gas chromatography techiques have also been used by others to assess the spontaneous liberation of CO from CO-RMs in solution. In parallel, we used bioassays such as relaxation of aortic vessels and inhibition of the inflammatory response in macrophages to assess the CO-mediated pharmacological effects of CO-RMs 12 57 91. In most cases we found a very good correlation between the rate and mode of CO release by CO-RMs and their effect on aortic ring relaxation. The results from several studies have also enabled us to propose that the chemical structure of metal carbonyls CO-RMs might facilitate the direct transfer of CO from CO-RMs to the intracellular target(s) as it appears that the release of CO from certain metal carbonyls (i.e. CORM-2 and CORM-3) requires an acceptor 17 44 92. This may enhance selectivity for the action of CO from metal carbonyls and the difference with CO gas applied exogenously would be that its diffusion into cells might be limited or hampered by the encounter of many proteins potentially able to bind CO, including the prototypic intracellular target(s) 17 77. Although this concept needs to be substantiated, results on the bactericidal effects of CO-RMs (see above) and an interesting article published recently reports data in this direction. Wang and colleagues have developed a genetically encoded fluorescent probe, which is capable of selectively detecting CO inside living cell 93. The probe, named COSer for CO sensor, consists of a permuted yellow fluorescent protein inserted into the regulatory domain of the bacterial protein CooA, a heme-dependent transcription factor known to bind CO with high affinity and selectivity. It was found that the fluorescent intensity of HeLa cells transfected with COSer increased after addition of 5 μM CO gas and a higher response was obtained with 10 μM. Interestingly, the fluorescence intensity was even stronger in cells treated with CORM-2 as a very significant response was obtained with only 1 μM CORM-2 and to obtain a given fluorescence intensity, more CO gas was needed with COSer-transfected cells than with the purified probe. These findings led the authors to state that CORM-2 provided an alternative and more controllable method for CO delivery to cells and could have possibly reduced the difficulty they encountered in getting CO into cells by using simple CO solutions. Similar findings were recently obtained by Michel and co-workers, who have synthesized a palladium-based fluorescent probe that is capable of detecting CO with high selectivity both in aqueous solutions and in living cells. Notably, CORM-3 was used in their experiments as the source of CO revealing that, unlike CO gas, concentrations as low as 1 μM CORM-3 were sufficient to trigger fluorescence in cells loaded with the palladium probe 94.

The use of the MbCO assay to assess the rate and amount of CO liberated by CO-RMs has been recently questioned 95. In our experiments we showed that while MbCO is promptly formed after addition of CORM-2 or CORM-3 to a solution containing reduced Mb, a sensitive CO electrode failed to detect any CO upon addition of these two CO-RMs 17. However, CO release from CORM-A1, a boranocarbonate, is detected by the Mb assay and by the electrode with comparable results, indicating the spontaneous liberation of CO from the compound. McLean and colleagues have shown that in the case of CORM-3 and CORM-2 liberation of CO and the consequent formation of MbCO is facilitated by dithionite, which is usually added in excess to the assay for keeping Mb in a reduced state 95. The authors concluded that the MbCO assay should be abandoned and propose the use of hemoglobin (Hb) since it binds CO with a much greater affinity than oxygen and does not require deoxygenation by dithionite. We believe these data indicate that dithionite and other sulphite can accelerate the release of CO from CO-RMs and that rates of CO release obtained with the MbCO assay should be interpreted cautiously, but we would like to add few important considerations still in favour of the MbCO assay. First, the results by McLean and colleagues seem to imply that liberation (or transfer) of CO from CORM-2 and CORM-3 to a prototypic target (i.e. Mb) cannot occur in the presence of a deoxygenated reduced heme but is triggered only by interaction with anions such as sulphites. That this is not the case is elegantly described by Obirai and colleagues in an interesting report published few years ago revealing quite the opposite as CORM-2 was demonstrated to directly transfer CO to a heme(FeII)/heme(FeIII) redox couple 92. Using a cyclic voltammetry method, the authors proved that when CORM-2 is added to an argon deaerated phosphate buffer solution containing an electrode coated with the heme redox couple but in complete absence of dithionite, a heme(FeII)-CO complex is formed. Secondly, the determination of the rate of CO liberation from CO-RMs by using an in vitro biochemical assay is rather approximate and we always judged it best to interpret our data on CO liberation using a combination of approaches as these compounds are designed for their possible therapeutic use in vivo. That is the reason why in our studies on the characterization of CO-RMs we always coupled the quantification of CO liberation in vitro with data obtained using bioassays that reflect more closely the behaviour of these compounds in complex biological systems. For instance, despite the fact that CORM-2 and CORM-3 are stable compounds in solution and may not liberate CO spontaneously, they still cause a rapid relaxation in isolated vessels and hypotension in animals suggesting that these compounds are fast CO releasers in vivo in line with the MbCO assay data. The bioactive effects mediated by the rapid CO release from these two CO-RMs have been corroborated by using pharmacological tools (i.e. inactive CO-RMs or CO-RMs depleted of CO) 15 57 91 or by comparison with compounds that release CO much slower in vitro and in vivo (i.e. CORM-A1 and CORM-371) 16 17. As a further example, we have recently employed the MbCO assay to determine that CORM-401, a manganese-containing CO-RM, liberates 3-4 CO per molecule 60. We found that the relaxation exerted by this molecule in aortic rings is approximately 3-fold more pronounced than that elicited by the same concentration of CORM-A1, which has a half-life similar to CORM-401 but liberates only 1 CO (unpublished results). Third, the use of oxygenated Hb instead of reduced Mb poses other relevant issues, such as the presence of 4 hemes and the co-operative effect of CO binding to the hemes, which will make it more difficult to quantify the amount and kinetics of CO released. These considerations, together with the results of the MbCO assay and the recent findings with the fluorescent probes reported above, strongly indicate that the release of CO from CORM-2 and CORM-3 occurs when the metal carbonyl is in the vicinity of a reduced iron acceptor (MbFe(II) or heme(II)). The results have also important implications on the efficacy of metal carbonyl CO-RMs in delivering CO to prototypic intracellular targets.

Conclusions

Although the initial discovery of CO-RMs took place a decade ago 12, it is intriguing that many diverse and novel pharmacological actions are being discovered for these compounds. The studies conducted so far reflect a real effort to understand the biochemical mechanisms that mediate the beneficial effects of CO-RMs. The emerging scenario is that, while CO-RMs mainly affect cellular functions via the liberation of CO, the molecules may facilitate or modulate other concomitant reactions involving redox and metal-sensitive pathways. Importantly, the chemical reactivity of metal carbonyl complexes-based CO-RMs may be enriching, rather than diminishing, their positive actions suggesting that a critical assessment of CO-RMs’ behaviour in biological environment (bioassays) must always be evaluated in parallel to their ability to release CO. In view of the growing importance of inflammatory components in initiating and modulating pathological processes, we have focused here on how CO-RMs modulate the inflammatory response as consistent and converging data point to their interesting anti-inflammatory activity. As new CO-RMs with a multiplicity of chemical properties and reactivity are being synthesized and tested in biological models, we may in the future uncover novel promising applications for this unique class of compounds.

Abbreviations

CLP: Cecal ligation and puncture; CO: Carbon monoxide; CO-RMs: Carbon monoxide-releasing molecules; CO: Carbon monoxide; CORM-2: [Ru(CO)3Cl2]2) also known as tricarbonyldichlororuthenium(II) dimer; CORM-3: Ru(CO)3Cl(glycinate) also known as tricarbonylchloro(glycinato)ruthenium(II); CORM-371: [Me4N][Mn(CO)4(thioacetate)2]; CORM-A1: NaH3BCOOH, also known as sodium boranocarbonates; COX-2: Cyclooxygenase-2; Hb: Hemoglobin; (HMOX1): Heme oxygenase-1; ICAM-1: Intercellular adhesion molecule 1; IL: Interleukin; IL-1β: Interleukin 1 beta; iNOS: Inducible nitric oxide synthase or NOS(III); LPS: Lypopolysaccharide; MAPKs: Mitogen activated protein kinases; Mb: Myoglobin; MbCO: Carbon monoxy myoglobin; MMP: Matrix metallo proteinase; MPO: Myeloperoxidase; NO: Nitric oxide; Nfkβ: Nuclear factor kappa beta; O2: Superoxide anion; PGE2: Prostaglandin E2; PMNs: Polymorphonuclear neutrophils; ROS: Reactive oxygen species; TNF-α: Tumor necrosis factor alpha.

Competing interests

Dr. Roberto Motterlini was founder and scientific director of hemoCORM (2004-2008), holds patents on the CO-RMs technology and is a shareholder of Alfama.

Authors’ contribution

RM and RF wrote the manuscript. BH drafted an earlier version of the manuscript and helped with the outline of Table 1. All authors have read and approved the manuscript.

Acknowledgements

We would like to thank the AREMCAR Foundation for financial support. The authors would also like to thank Prof. Brian Mann (University of Sheffield, United Kingdom) and all the former colleagues at Northwick Park Institute for Medical Research (Harrow, United Kingdom) for their contribution and support.

<p>Microsomal heme oxygenase. Characterization of the enzyme</p>TenhunenRMarverHSSchmidRJ Biol Chem1969244638863944390967<p>Heme oxygenase: function, multiplicity, regulatory mechanisms, and clinical applications</p>MainesMDFASEB J19882255725683290025<p>The regulation of heme turnover and carbon monoxide biosynthesis in cultured primary rat olfactory receptor neurons</p>IngiTChiangGRonnettGVJ Neurosci199616562156288795618<p>Heme oxygenase-1-derived carbon monoxide contributes to the suppression of acute hypertensive responses in vivo</p>MotterliniRGonzalesAForestiRClarkJEGreenCJWinslowRMCirc Res19988356857710.1161/01.RES.83.5.5689734480<p>Induction of heme oxygenase: a general response to oxidant stress in cultured mammalian cells</p>ApplegateLALuscherPTyrrellRMCancer Res1991519749781988141<p>Rapid induction of heme oxygenase-1 mRNA and protein by hyperthermia in rat brain: heme oxygenase-2 is not a heat shock protein</p>EwingJFMainesMDProc Natl Acad Sci USA1991885364536810.1073/pnas.88.12.5364518732052613<p>Bilirubin is an antioxidant of possible physiological importance</p>StockerRYamamotoYMcDonaghAFGlazerANAmesBNScience19872351043104610.1126/science.30298643029864<p>Carbon monoxide has anti-inflammatory effects involving the mitogen-activated protein kinase pathway</p>OtterbeinLEBachFHAlamJSoaresMTao LuHWyskMDavisRJFlavellRAChoiAMNature Med2000642242810.1038/7468010742149<p>The therapeutic potential of carbon monoxide</p>MotterliniROtterbeinLENat Rev Drug Discov2010972874310.1038/nrd322820811383<p>Carbon monoxide in biology and medicine</p>RyterSWOtterbeinLEBioEssays20042627028010.1002/bies.2000514988928<p>Carbon monoxide: innovative anti-inflammatory properties of an age-old gas molecule</p>OtterbeinLEAntioxid Redox Signal2002430931910.1089/15230860275366636112006182<p>Carbon monoxide-releasing molecules: characterization of biochemical and vascular activities</p>MotterliniRClarkJEForestiRSarathchandraPMannBEGreenCJCirc Res200290E17E2410.1161/hh0202.10453011834719<p>Metal carbonyls: a new class of pharmaceuticals?</p>JohnsonTRMannBEClarkJEForestiRGreenCJMotterliniRAngew Chem Int Ed Engl2003423722372910.1002/anie.20030163412923835<p>Bioactivity and pharmacological actions of carbon monoxide-releasing molecules</p>MotterliniRMannBEJohnsonTRClarkJEForestiRGreenCJCurr Pharmacol Design200392525253910.2174/1381612033453785<p>Cardioprotective actions by a water-soluble carbon monoxide-releasing molecule</p>ClarkJENaughtonPShureySGreenCJJohnsonTRMannBEForestiRMotterliniRCirc Res200393e2e810.1161/01.RES.0000084381.86567.0812842916<p>CORM-A1: a new pharmacologically active carbon monoxide-releasing molecule</p>MotterliniRSawlePBainsSHammadJAlbertoRForestiRGreenCJFASEB J20051928428615556971<p>Differential antibacterial activity against pseudomonas aeruginosa by carbon monoxide-releasing molecules</p>DesmardMForestiRMorinDDagoussatMBerdeauxADenamurECrookSHMannBEScapensDMontraversPBoczkowskiJMotterliniRAntioxid Redox Signal20111615316321864022<p>Antithrombotic properties of water-soluble carbon monoxide-releasing molecules</p>KramkowskiKLeszczynskaAMogielnickiAChlopickiSFedorowiczAGrochalEMannBBrzoskaTUranoTMotterliniRBuczkoWArterioscler Thromb Vasc Biol2012322149215710.1161/ATVBAHA.112.25398922772756<p>CORM-3, a carbon monoxide-releasing molecule, alters the inflammatory response and reduces brain damage in a rat model of hemorrhagic stroke</p>YabluchanskiyASawlePHomer-VanniasinkamSGreenCJForestiRMotterliniRCrit Care Med20124054455210.1097/CCM.0b013e31822f0d6421926571<p>Enhanced induction of heme oxygenase-1 suppresses thrombus formation and affects the protein C system in sepsis</p>FeiDMengXZhaoMKangKTanGPanSLuoYLiuWNanCJiangHKrissansenGWZhaoMSunXTransl Res20121599910910.1016/j.trsl.2011.10.00922243794<p>Downregulation of the inflammatory response by CORM-3 results in protective effects in a model of postmenopausal arthritis</p>IbanezLAlcarazMJMaicasNGuedeDCaeiroJRMotterliniRFerrandizMLCalcif Tissue Int201291698010.1007/s00223-012-9612-722644323<p>Carbon monoxide (CO)-releasing molecule-derived CO regulates tissue factor and plasminogen activator inhibitor type 1 in human endothelial cells</p>MaruyamaKMorishitaEYunoTSekiyaAAsakuraHOhtakeSYachieAThromb Res2012130e188e19310.1016/j.thromres.2012.07.00222819264<p>Carbon monoxide reduces neuropathic pain and spinal microglial activation by inhibiting nitric oxide synthesis in mice</p>HerveraALeanezSNegreteRMotterliniRPolOPLoS One20127e4369310.1371/journal.pone.0043693342550722928017<p>The carbon monoxide releasing molecule (CORM-3) inhibits expression of vascular cell adhesion molecule-1 and E-selectin independently of haem oxygenase-1 expression</p>SongHBergstrasserCRafatNHogerSSchmidtMEndresNGoebelerMHillebrandsJLBrigelius-FloheRBanningABeckGLoeselRYardBABr J Pharmacol200915776978010.1111/j.1476-5381.2009.00215.x272126219422386<p>Preconditioning of carbon monoxide releasing molecule-derived CO attenuates LPS-induced activation of HUVEC</p>SunBZouXChenYZhangPShiGInt J Biol Sci20084270278251983718726003<p>Inhibition of VCAM-1 expression in endothelial cells by CORM-3: the role of the ubiquitin-proteasome system, p38, and mitochondrial respiration</p>BergstraesserCHoegerSSongHErmantrautLHottenrotMCzymaiTSchmidtMGoebelerMPoneliesNStichCLoeselRMolemaGSeelenMvan SonWYardBARafatNFree Radic Biol Med20125279480210.1016/j.freeradbiomed.2011.11.03522210380<p>Carbon monoxide-releasing molecules protect against ischemia-reperfusion injury during kidney transplantation</p>CaumartinYStephenJDengJPLianDLanZLiuWGarciaBJevnikarAMWangHCepinskasGLukePPKidney Int2011791080108910.1038/ki.2010.54221270767<p>Carbon monoxide liberated from carbon monoxide-releasing molecule exerts an anti-inflammatory effect on dextran sulfate sodium-induced colitis in mice</p>TakagiTNaitoYUchiyamaKSuzukiTHirataIMizushimaKTsuboiHHayashiNHandaOIshikawaTYagiNKokuraSIchikawaHYoshikawaTDig Dis Sci2011561663167110.1007/s10620-010-1484-y21086163<p>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</p>ChungJShinDYZhengMJoeYPaeHORyterSWChungHTMol Immunol2011481793179910.1016/j.molimm.2011.05.01421640381<p>Wound healing activity of carbon monoxide liberated from CO-releasing molecule (CO-RM)</p>AhangerAAPrawezSKumarDPrasadRAmarpalTandanSKKumarDNaunyn Schmiedebergs Arch Pharmacol20113849310210.1007/s00210-011-0653-721584760<p>Carbon monoxide-releasing molecule-2 (CORM-2) attenuates acute hepatic ischemia reperfusion injury in rats</p>WeiYChenPde BruynMZhangWBremerEHelfrichWBMC Gastroenterol2010104210.1186/1471-230X-10-42287360120444253<p>Effects of carbon monoxide releasing molecule-liberated CO on severe acute pancreatitis in rats</p>ChenPSunBChenHWangGPanSKongRBaiXWangSCytokine201049152310.1016/j.cyto.2009.09.01319900821<p>The CO-releasing molecule CORM-3 protects against articular degradation in the K/BxN serum transfer arthritis model</p>MaicasNFerrandizMLDevesaIMotterliniRKoendersMIvan den BergWBAlcarazMJEur J Pharmacol201063418419110.1016/j.ejphar.2010.02.02820184873<p>Carbon monoxide liberated from CO-releasing molecule (CORM-2) attenuates ischemia/reperfusion (I/R)-induced inflammation in the small intestine</p>KatadaKBihariAMizuguchiSYoshidaNYoshikawaTFraserDDPotterRFCepinskasGInflammation2010339210010.1007/s10753-009-9162-y19842024<p>Carbon monoxide induces cyclooxygenase-2 expression through MAPKs and PKG in phagocytes</p>LinLCHoFMYenSJWuPYHungLFHuangWJLiangYCInt Immunopharmacol2010101520152510.1016/j.intimp.2010.08.02620840837<p>Carbon monoxide rescues mice from lethal sepsis by supporting mitochondrial energetic metabolism and activating mitochondrial biogenesis</p>LancelSHassounSMFavoryRDecosterBMotterliniRNeviereRJ Pharmacol Exp Ther20091329641648<p>CO-releasing molecule (CORM-3)-derived CO modulates neutrophil (PMN) migration across vascular endothelium by reducing the levels of cell surface-bound elastase</p>MizuguchiSStephenJDencev-BihariRMarkovicNSuehiroSCaprettaFPotterRFCepinskasGAm J Physiol Heart Circ Physiol2009297H920H92910.1152/ajpheart.00305.200919561312<p>Heme-oxygenase-1 induction and carbon monoxide-releasing molecule inhibit lipopolysaccharide (LPS)-induced high-mobility group box 1 release in vitro and improve survival of mice in LPS- and cecal ligation and puncture-induced sepsis model in vivo</p>TsoyiKLeeTYLeeYSKimHJSeoHGLeeJHChangKCMol Pharmacol20097617318210.1124/mol.109.05513719366789<p>Carbon monoxide liberated from carbon monoxide-releasing molecule CORM-2 attenuates inflammation in the liver of septic mice</p>CepinskasGKatadaKBihariAPotterRFAm J Physiol Gastrointest Liver Physiol2008294G184G19117991708<p>Nox4 NADPH oxidase mediates oxidative stress and apoptosis caused by TNF-α in cerebral vascular endothelial cells</p>BasuroySBhattacharyaSLefflerCWParfenovaHAm J Physiol Cell Physiol2009296C422C432266026219118162<p>Water-soluble CO-releasing molecules (CO-RMs) reduce the development of postoperative ileus via modulation of MAPK/HO-1 signaling and reduction of oxidative stress</p>De BackerOElinckEBlanckaertBLeybaertLMotterliniRLefebvreRAGut20095834735610.1136/gut.2008.15548119022916<p>CO liberated from CORM-2 modulates the inflammatory response in the liver of thermally injured mice</p>SunBWSunYSunZWChenXWorld J Gastroenterol20081454755310.3748/wjg.13.6127268114518203286<p>Carbon liberated from CO-releasing molecules attenuates leukocyte infiltration in the small intestine of thermally injured mice</p>SunBWJinQSunYSunZWChenXChenZYCepinskasGWorld J Gastroenterol2007136183619010.3748/wjg.13.618318069757<p>Carbon monoxide-releasing antibacterial molecules target respiration and global transcriptional regulators</p>DavidgeKSSanguinettiGYeeCHCoxAGMcLeodCWMonkCEMannBEMotterliniRPooleRKJ Biol Chem20092844516452419091747<p>Reactive oxygen species mediate bactericidal killing elicited by carbon monoxide-releasing molecules</p>TavaresAFTeixeiraMRomaoCCSeixasJDNobreLSSaraivaLMJ Biol Chem2011286267082671710.1074/jbc.M111.255752314363421646348<p>The carbon monoxide releasing molecule CORM-2 attenuates Pseudomonas aeruginosa biofilm formation</p>MurrayTSOkegbeCGaoYKazmierczakBIMotterliniRDietrichLEBrusciaEMPLoS One20127e3549910.1371/journal.pone.0035499333852322563385<p>Antimicrobial action of carbon monoxide-releasing compounds</p>NobreLSSeixasJDRomaoCCSaraivaLMAntimicrob Agents Chemother2007514303430710.1128/AAC.00802-07216797417923486<p>High mobility group Box 1 contributes to lethality of endotoxemia in heme oxygenase-1 deficient mice</p>TakamiyaRHungCCHallSRFukunagaKNagaishiTMaenoTOwenCMaciasAAFredenburghLEIshizakaABlumbergRSBaronRMPerrellaMAAm J Respir Cell Mol Biol200841129135271590219097991<p>Treatment with a CO-releasing molecule (CORM-3) reduces joint inflammation and erosion in murine collagen-induced arthritis</p>FerrandizMLMaicasNGarcia-ArnandisITerencioMCMotterliniRDevesaIJoostenLAvan den BergWBAlcarazMJAnn Rheum Dis2008671211121718063671<p>A carbon monoxide-releasing molecule (CORM-3) abrogates polymorphonuclear granulocyte-induced activation of endothelial cells and mast cells</p>MasiniEVannacciAFailliPMastroianniRGianniniLVinciMCUlivaCMotterliniRMannaioniPFFASEB J2008223380338810.1096/fj.08-10711018556460<p>The CO-releasing molecule CORM-2 is a novel regulator of the inflammatory process in osteoarthritic chondrocytes</p>GuillenMIMegiasJCleriguesVGomarFAlcarazMJRheumatology (Oxford)2008471323132810.1093/rheumatology/ken264<p>The carbon monoxide-releasing molecule CORM-2 inhibits the inflammatory response induced by cytokines in Caco-2 cells</p>MegiasJBusserollesJAlcarazMJBr J Pharmacol2007150977986201391317339836<p>The carbon monoxide-releasing molecule tricarbonyldichlororuthenium(II) dimer protects human osteoarthritic chondrocytes and cartilage from the catabolic actions of interleukin-1beta</p>MegiasJGuillenMIBruAGomarFAlcarazMJJ Pharmacol Exp Ther2008325566110.1124/jpet.107.13465018195133<p>Carbon monoxide-releasing molecules modulate leukocyte-endothelial interactions under flow</p>UrquhartPRosignoliGCooperDMotterliniRPerrettiMJ Pharmacol Exp Ther200732165666210.1124/jpet.106.11721817289832<p>Modulation of thrombin-induced neuroinflammation in BV-2 microglia by a carbon monoxide-releasing molecule (CORM-3)</p>Bani-HaniMGGreensteinDMannBEGreenCJMotterliniRJ Pharmacol Exp Ther20063181315132210.1124/jpet.106.10472916772536<p>A carbon monoxide-releasing molecule (CORM-3) attenuates lipopolysaccharide- and interferon-gamma-induced inflammation in microglia</p>Bani-HaniMGGreensteinDMannBEGreenCJMotterliniRPharmacol Rep200658Suppl13214417332683<p>Carbon monoxide-releasing molecules (CO-RMs) attenuate the inflammatory response elicited by lipopolysaccharide in RAW264.7 murine macrophages</p>SawlePForestiRMannBEJohnsonTRGreenCJMotterliniRBr J Pharmacol200514580081010.1038/sj.bjp.0706241157619515880142<p>Bioactive properties of iron-containing carbon monoxide-releasing molecules (CO-RMs)</p>SawlePHammadJFairlambIJMoultonBO'BrienCTLynamJMDuhme-KlairAKForestiRMotterliniRJ Pharmacol Exp Ther200631840341010.1124/jpet.106.10175816603670<p>Iron indenyl carbonyl compounds: CO-releasing molecules</p>HewisonLCrookSHJohnsonTRMannBEAdamsHPlantSESawlePMotterliniRDalton Trans2010398967897510.1039/c0dt00203h20714615<p>[Mn(CO)4{S2CNMe(CH2CO2H)}], a new water-soluble CO-releasing molecule</p>CrookSHMannBEMeijerJAHMAdamsHSawlePScapensDMotterliniRDalton Trans2011404230423510.1039/c1dt10125k21403944<p>Gaseotransmitters: new frontiers for translational science</p>SzaboCSci Transl Med2010259ps5410.1126/scitranslmed.3000721303860521106939<p>Ruthenium imidazole oxime carbonyls and their activities as CO-releasing molecules</p>OresmaaLTarvainenHMachalKHaukkaMDalton Trans201241111701117510.1039/c2dt31002c22868452<p>Photoactivated biological activity of transition-metal complexes</p>SchatzschneiderUEur J Inorg Chem20104114511467<p>Silicium dioxide nanoparticles as carriers for photoactivatable CO-releasing molecules (PhotoCORMs)</p>DordelmannGPfeifferHBirknerASchatzschneiderUInorg Chem2011504362436710.1021/ic102419721506524<p>Acyloxybutadiene iron tricarbonyl complexes as enzyme-triggered CO-releasing molecules (ET-CORMs)</p>RomanskiSKrausBSchatzschneiderUNeudorflJMAmslingerSSchmalzHGAngew Chem Int Ed Engl2011502392239621351362<p>Carbon monoxide-releasing micelles for immunotherapy</p>HasegawaUvan der VliesAJSimeoniEWandreyCHubbellJAJ Am Chem Soc2010132182731828010.1021/ja107502521128648<p>CO releasing properties and cytoprotective effect of cis-trans-[Re(II)(CO)2Br2L2]n complexes</p>ZobiFDegondaASchaubMCBogdanovaAYInorg Chem2010497313732210.1021/ic100458j20690741<p>Redox signaling in inflammation: interactions of endogenous electrophiles and mitochondria in cardiovascular disease</p>KoenitzerJRFreemanBAAnn N Y Acad Sci20101203455210.1111/j.1749-6632.2010.05559.x20716282<p>Heme oxygenase: a novel target for the modulation of inflammatory response</p>WillisDMooreARFrederickRWilloughbyDANature Med19962879010.1038/nm0196-878564848<p>Oxidative stress causes enhanced endothelial cell injury in human heme oxygenase-1 deficiency</p>YachieANiidaYWadaTIgarashiNKanedaHTomaTOhtaKKasaharaYKoizumiSJ Clin Invest199910312913510.1172/JCI41654078589884342<p>Heme oxygenase 1 is required for mammalian iron reutilization</p>PossKDTonegawaSProc Natl Acad Sci USA199794109191092410.1073/pnas.94.20.10919235319380735<p>Biliverdin rescues the HO-2 null mouse phenotype of unresolved chronic inflammation following corneal epithelial injury</p>BellnerLWolsteinJPatilKADunnMWLaniado-SchwartzmanMInvest Ophthalmol Vis Sci2011523246325310.1167/iovs.10-6219310902621345995<p>Biliverdin administration protects against endotoxin-induced acute lung injury in rats</p>Sarady-AndrewsJKLiuFGalloDNakaoAOverhausMOllingerRChoiAMOtterbeinLEAm J Physiol Lung Cell Mol Physiol2005289L1131L113710.1152/ajplung.00458.200416155084<p>Heme oxygenase-1/carbon monoxide: from basic science to therapeutic applications</p>RyterSWAlamJChoiAMPhysiol Rev20068658365010.1152/physrev.00011.200516601269<p>Role of CO-releasing molecules liberated CO in attenuating leukocytes sequestration and inflammatory responses in the lung of thermally injured mice</p>SunBSunHLiuCShenJChenZChenXJ Surg Res200713912813510.1016/j.jss.2006.08.03217292406<p>Carbon monoxide improves cardiac function and mitochondrial population quality in a mouse model of metabolic syndrome</p>LancelSMontaigneDMarechalXMarciniakCHassounSMDecosterBBallotCBlazejewskiCCorseauxDLescureBMotterliniRNeviereRPLoS One20127e4183610.1371/journal.pone.0041836341156922870253<p>Interaction of carbon monoxide with transition metals: evolutionary insights into drug target discovery</p>ForestiRMotterliniRCurr Drug Targets2010111595160410.2174/138945011100901159520704543<p>Mitochondrial respiratory chain and NAD(P)H oxidase are targets for the antiproliferative effect of carbon monoxide in human airway smooth muscle</p>TailleCEl-BennaJLanoneSBoczkowskiJMotterliniRJ Biol Chem2005280253502536010.1074/jbc.M50351220015863496<p>A carbon monoxide-releasing molecule (CORM-3) uncouples mitochondrial respiration and modulates the production of reactive oxygen species</p>Lo IaconoLBoczkowskiJZiniRSalouageIBerdeauxAMotterliniRMorinDFree Rad Biol Med2011501556156410.1016/j.freeradbiomed.2011.02.03321382478<p>CO from enhanced HO activity or from CORM-2 inhibits both O2- and NO production and downregulates HO-1 expression in LPS-stimulated macrophages</p>SrisookKHanSSChoiHSLiMHUedaHKimCChaYNBiochem Pharmacol20067130731810.1016/j.bcp.2005.10.04216329999<p>Heme Oxygenase-2 provides endogenous protection against oxidative stress and apoptosis caused by TNF-α in cerebral vascular endothelial cells</p>BasuroySBhattacharyaSTcheranovaDQuYReganRFLefflerCWParfenovaHAm J Physiol Cell Physiol2006291C897C90810.1152/ajpcell.00032.200616822952<p>TNF-alpha/cycloheximide-induced oxidative stress and apoptosis in murine intestinal epithelial MODE-K cells</p>BabuDSoenenSJRaemdonckKLeclercqGDe BackerOMotterliniRLefebvreRACurr Pharm Des2012184414442510.2174/13816121280248129122721505<p>Essential role of induced nitric oxide in the initiation of the inflammatory response after hemorrhagic shock</p>HierholzerCHarbrechtBMenezesJMKaneJMacMickingJNathanCFPeitzmanABBilliarTRTweardyDJJ Exp Med199818791792810.1084/jem.187.6.91722121859500794<p>Stopped-flow analysis of CO and NO binding to inducible nitric oxide synthase</p>AbuSoudHMWuCQGhoshDKStuehrDJBiochemistry1998373777378610.1021/bi972398q9521697<p>Astrocytes induce hemeoxygenase-1 expression in microglia: a feasible mechanism for preventing excessive brain inflammation</p>MinKJYangMSKimSUJouIJoeEHJ Neurosci2006261880188710.1523/JNEUROSCI.3696-05.200616467537<p>Heme oxygenase-1-derived carbon monoxide enhances the host defense response to microbial sepsis in mice</p>ChungSWLiuXMaciasAABaronRMPerrellaMAJ Clin Invest200811823924710.1172/JCI32730210448018060048<p>A carbon monoxide-releasing molecule (CORM-3) exerts bactericidal activity against Pseudomonas aeruginosa and improves survival in an animal model of bacteraemia</p>DesmardMDavidgeKSBouvetOMorinDRouxDForestiRRicardJDDenamurEPooleRKMontraversPMotterliniRBoczkowskiJFASEB J2009231023103110.1096/fj.08-12280419095732<p>A role for reactive oxygen species in the antibacterial properties of carbon monoxide-releasing molecules</p>TavaresAFNobreLSSaraivaLMFEMS Microbiol Lett201233611010.1111/j.1574-6968.2012.02633.x22774863<p>Antibacterial effects of carbon monoxide</p>WilsonJLJesseHEPooleRKDavidgeKSCurr Pharm Biotechnol20121376076822201612<p>Exploring the antimicrobial action of a carbon monoxide-releasing compound through whole-genome transcription profiling of Escherichia coli</p>NobreLSAl ShahrourFDopazoJSaraivaLMMicrobiology200915581382410.1099/mic.0.023911-019246752<p>Vasoactive properties of CORM-3, a novel water-soluble carbon monoxide-releasing molecule</p>ForestiRHammadJClarkJEJohnsonRAMannBEFriebeAGreenCJMotterliniRBr J Pharmacol200414245346010.1038/sj.bjp.0705825157497915148243<p>UV-visible and electrochemical monitoring of carbon monoxide release by donor complexes to myoglobin solutions and to electrodes modified with films containing hemin</p>ObiraiJCHamadiSIthurbideAWartelleCNyokongTZagalJTopSBediouiFElectroanalyis2006181689169510.1002/elan.200603571<p>A selective fluorescent probe for carbon monoxide imaging in living cells</p>WangJKarpusJZhaoBSLuoZChenPRHeCAngew Chem Int Ed Engl2012519652965610.1002/anie.20120368422930547<p>A reaction-based fluorescent probe for selective imaging of carbon monoxide in living cells using a palladium-mediated carbonylation</p>MichelBWLippertARChangCJJ Am Chem Soc2012134156681567110.1021/ja307017b22970765<p>Sulfite species enhance carbon monoxide release from CO-releasing molecules: implications for the deoxymyoglobin assay of activity</p>McLeanSMannBEPooleRKAnal Biochem2012427364010.1016/j.ab.2012.04.02622561917