Skip to main content
American Journal of Physiology - Lung Cellular and Molecular Physiology logoLink to American Journal of Physiology - Lung Cellular and Molecular Physiology
. 2015 Oct 23;309(12):L1387–L1393. doi: 10.1152/ajplung.00311.2015

Carbon monoxide in the treatment of sepsis

Kiichi Nakahira 1,2, Augustine M K Choi 1,2,
PMCID: PMC4683310  PMID: 26498251

Abstract

Carbon monoxide (CO), a low-molecular-weight gas, is endogenously produced in the body as a product of heme degradation catalyzed by heme oxygenase (HO) enzymes. As the beneficial roles of HO system have been elucidated in vitro and in vivo, CO itself has also been reported as a potent cytoprotective molecule. Whereas CO represents a toxic inhalation hazard at high concentration, low-dose exogenous CO treatment (∼250–500 parts per million) demonstrates protective functions including but not limited to the anti-inflammatory and antiapoptotic effects in preclinical models of human diseases. Of note, CO exposure confers protection in animal models of sepsis by inhibiting inflammatory responses and also enhancing bacterial phagocytosis in leukocytes. These unique functions of CO including both dampening inflammation and promoting host defense mechanism are mediated by multiple pathways such as autophagy induction or biosynthesis of specialized proresolving lipid mediators. We suggest that CO gas may represent a novel therapy for patients with sepsis.

Keywords: clinical trial, sepsis, carbon monoxide


sepsis, defined as suspected or proven infection accompanied by a systemic inflammatory response syndrome, remains one of the leading causes of death and a major challenge in intensive care units (1, 54). There are more than 750,000 cases of sepsis every year in the United States (37). The number of sepsis cases per year is increasing as a result of the aging population, the increased longevity of people with chronic diseases, the spread of antibiotic-resistant organisms, and broader use of immunosuppressive and chemotherapeutic agents (21, 54). A recent study reports that sepsis contributes to one in every two to three deaths in hospitals (35).

The pathophysiology of sepsis is complex, with interactions between the infecting microorganism and the host responses including systemic immune, inflammatory, and coagulation responses (1, 54, 68). Furthermore, excess host responses to infections often trigger tissue/organ injuries and subsequent secondary infection (1). The complex heterogeneous and not-fully-understood pathogenesis in septic patients remains a big hurdle to overcome sepsis (1, 36, 68), with many unfavorable outcomes in clinical trials (36). The recent failure and withdrawing of Xigris (activated protein C) from the market represents one example of the difficulties of designing effective drugs or interventions for patients with sepsis (2, 36, 53, 77).

After discovery of nitric oxide (NO) as a cellular signaling molecule involved in various physiological and pathological processes (23, 24, 40), other endogenously produced gaseous molecules such as CO and hydrogen sulfide (H2S) have been studied as potential therapeutics in human diseases (16, 65, 76). In particular, the beneficial roles of low-dose carbon monoxide (CO) have been reported in a number of studies during the last 15 years (43, 55, 60). In this Perspectives, we highlight the cytoprotective roles of CO with focus on the beneficial function of CO in sepsis and discuss the potential therapeutic use of CO for treatment of sepsis in humans.

CO as a Potent Cytoprotective Molecule

CO is a ubiquitous environmental product of organic combustion process, including the burning of wood, coal, gas, and tobacco (72). Indoor ambient levels of CO range are from 0.5 to 5 ppm. CO is also physiologically produced in cells and tissues as the byproduct of heme catalytic activity of heme oxygenase (HO) enzymes (55, 60). The rate of CO generation in the human body may substantially change, when cells or tissues increase the expression of inducible HO enzyme (i.e., HO-1) in response to various oxidative stresses (55, 60). HO-1 is the rate-limiting enzyme in the degradation of heme to generate CO, iron, and biliverdin-IXa, which is converted to bilirubin-IXa (55, 60). Beneficial roles of the HO enzyme system have been studied in various disease models (18, 51, 55, 59, 60). With the ever-increasing amount of experimental evidence demonstrating the beneficial roles of HO, much interest has been generated by both basic and clinical/translational investigators whether direct administration of such end-products may serve as a therapeutic approach in human diseases. Despite the notorious pharmacological effect of CO (i.e., the high affinity for hemoglobin and the resultant displacement of oxygen) (17, 52), many studies have demonstrated the protective roles of inhaled CO, when administered at low concentration, in various disease models including hyperoxia-induced acute lung injury (ALI), mechanical ventilation-induced ALI, ischemia/reperfusion (I/R) injury, vascular injury, graft rejection (43, 55, 60). Low-dose CO inhalation also improves the survival of septic rodents induced by cecal ligation and puncture (CLP), endotoxin, Enterococcus faecalis, or Escherichia coli (30, 42, 43, 74). Similar protective effects have also been observed in septic mice subjected to administration of chemical CO-donor compounds termed carbon monoxide releasing molecules (9, 13, 43). Of note, the protective roles of CO on sepsis is not limited to rodent models but also observed in large animal models such as swine and nonhuman primates subjected to LPS or Streptococcus pneumoniae administration (12, 15, 39).

Dampening inflammation.

One of the potent protective functions of CO in sepsis models is the anti-inflammatory effect. In 2000, Otterebein et al. (48) reported that CO inhalation (250 ppm) suppresses proinflammatory cytokine production such as TNF-α and increased anti-inflammatory cytokine production such as IL-10 in endotoxemia. Furthermore, CO inhibits proinflammatory signaling pathways activated by other pathogen-associated molecular patterns including Toll-like receptor (TLR) 2-, TLR5-, and TLR9-ligands (46). These reports suggest that the anti-inflammatory property of CO is effective on a wide range of microbial components. The general mechanisms of anti-inflammatory effects of CO include activation of p38 MAPK, suppression of translocation of TLRs, and enhanced PPAR-γ signaling (43, 55, 57, 58). A recent report has also demonstrated another new target of CO to dampen inflammation (25). Inflammasomes are cytosolic protein complexes that promote the cleavage of caspase-1 and secretion of IL-1β and IL-18 and are critically involved in the pathogenesis of various inflammatory diseases (20, 29). The levels of circulating IL-18 are increased in patients with acute respiratory distress syndrome (ARDS) and sepsis and are associated with the patients' mortality in medical intensive care unit (14). CO negatively regulates NLRP3-dependent inflammasome activation and IL-18 secretion (25). Thus the negative regulation of CO on IL-18 secretion may represent one key mechanism to ameliorate the pathogenesis of sepsis. Although a number of studies show the anti-inflammatory effect of CO on immune cells such as macrophages, it should be noted that CO also inhibits production of chemokine such as monocyte chemotactic protein 1 in type I alveolar epithelial cells (4). Thus CO can efficiently dampen systemic inflammatory responses by regulating immune responses of various types of cells.

Eliminating microbes.

Although regulating inflammation is an important arm to prevent multiorgan dysfunctions in sepsis, proper immune responses are critical host responses to eliminate microbes during infection (1, 54, 68). In this regard, CO can also regulate a key host defense mechanism in sepsis: promoting phagocytosis. CO promotes uptake of bacteria such as E. coli or E. faecalis by macrophages through activating autophagy (30) or redistribution of TLR4 on the cell surface (50). CO can also promote bacterial killing in macrophages by directly manipulating the metabolic pathways of microbes. Bacteria exposed with CO increase generation and secretion of ATP, in turn stimulating macrophage phagocytosis through activation of P2X7 receptor (74). Finally, recent reports demonstrate that CO promotes both bacterial killing and anti-inflammatory process mediated by biosynthesis of lipid mediators, critical molecules for maintaining tissue homeostasis. Emerging evidence indicates that cells/host can promote resolution of acute inflammation by activation of biosynthesis of specialized proresolving lipid mediators (SPMs) such as arachidonic acid-derived lipoxins or docosahexaenoic acid-derived D-series or E-series resolvins (RvDs or RvEs) (32, 61, 62). These SPMs terminate inflammation by inhibiting neutrophil infiltration to the inflaming sites and also enhancing macrophage phagocytosis, microbial clearance, and efflux to lymph nodes (32, 61, 62). The exogenous administration of SPMs such as resolvins improves the survival of septic mice subjected to bacterial infection (7, 67), and CO indeed upregulates phagocytosis by increasing biosynthesis of SPMs such as lipoxins and RvE in vivo and in vitro (8, 12, 63). Thus, unlike other immunomodulators used in previous clinical trials (e.g., targeting TNF-α or endotoxin), CO can exert its potent bacterial killing function and anti-inflammatory function through multiple mechanisms.

Regulating cell death and organ dysfunction.

In addition to immune responses, cell death also critically contributes to the pathogenesis of sepsis (1, 68). Released damage-associated molecular patterns (DAMPs) from dead cells of damaged tissues during sepsis can promote further immune responses (1, 27, 45). CO inhibits cell death in various cell types including epithelial cells or endothelial cells by regulating mitochondrial functions (31, 73). Mitochondrial dysfunction has been observed in septic patients (6, 19, 64, 70, 75, 78), and recent studies suggest that mitochondrial dysfunction might be associated with critically ill patients' outcome (28, 45, 47). For example, the levels of circulating cell-free mitochondrial DNA (mtDNA), a representative mitochondrial DAMP (45), are increased in patients with of sepsis (47). The elevated mtDNA levels predict an increased risk of death that persisted over up to 3.5 years of follow-up (47). Thus the inhibition of mitochondrial DAMPs release may likely to be a critical step to prevent sepsis-induced mortality. CO inhibits leakage of mtDNA from mitochondria by regulating mitochondrial membrane potential and mitochondrial reactive oxygen species generation (25). In addition, CO inhibits cell death in lung epithelial cells or kidney mesangial cells through upregulation of autophagy (26, 31). While autophagy is known as a cellular-protective mechanism, autophagy can also induce cell death in some oxidative stress models such as influenza A virus infection or sea water exposure (33, 44, 69). Although low-dose CO exposure may induce cell death, it should be noted that CO promotes resolution of acute inflammation in immune cells such as neutrophils and T cells by inducing cell death (8, 66). This pro-cell death function by CO may rather consequently inhibit tissue injuries triggered by inflammation. However, care may need to be taken for the cases with lung injuries in which autophagic cell death is involved. Finally, CO also exerts its protective effects in various organs or tissues including lung (12, 15, 30, 31, 34, 39), liver (30, 39, 81), heart (39), kidney (11, 26, 30, 39), and intestine (34, 41, 71)- important target organs of sepsis (Fig. 1). CO treatment can also improve the coagulation status with decreased disseminated intravascular coagulopathy (DIC) in a swine model of endotoxin challenge (39). Given the high mortality of sepsis with acute organ dysfunction and DIC, the prevention of organ/tissue dysfunction is an important strategy for treatment of sepsis (1, 54). Thus the protective effect of CO on multiple organs will be advantageous as an intervention for sepsis.

Fig. 1.

Fig. 1.

Protective roles of CO in various organs/cells during sepsis. Inhaled CO gas enters into the blood stream through alveolae. Most of CO in the blood is bound to hemoglobin as CO-Hb (∼80%) and the remaining CO is distributed into the tissues (10). The cellular concentration of CO is also influenced by the local partial pressure of both CO and oxygen (10). Distributed CO into organs/tissues exerts its various functions dependent on cell types. Inflammatory responses, invading microbes, and hypoxic condition during sepsis may lead to cellular dysfunction in various organs. CO regulates deteriorated cellular signaling and function to maintain cellular homeostasis. The cytoprotective effects of CO may differ among tissues/organs.

CO as a Therapeutic in Human Diseases

Although published works from the bench demonstrate CO as a potent “antiseptic” molecule, there are also important data from bedsides showing that the pathological conditions may increase production of CO in patients (22, 56). Exhaled CO (eCO) has been studied in patients as an inflammatory marker of diseases (3, 22, 56, 80) and may reflect increased production of systemic CO derived from HO induction in stressed pathophysiological conditions. The origin of eCO reflects a systemic elimination process of CO from the pulmonary circulation to the alveolae (22, 56). The levels of eCO are increased in patients who have systemic inflammatory responses or local inflammation (56). Of note, the levels of eCO are higher in mechanically ventilated septic patients, compared with ventilated patients without sepsis (79). More importantly, elevated eCO values on the first day of treatment are correlated with the probability of patient survival (79), suggesting the potentially critical roles of CO in septic patients. The first CO study in human was performed to investigate the effect of CO inhalation on inflammation during human endotoxemia (38). Each volunteer inhaled synthetic air (as placebo) and 500 ppm CO for 1 h in random order and received LPS intravenously after the inhalation. CO-Hb was increased from 1.2 to 7% by CO inhalation (38). In this study, the CO inhalation did not show significant effects on production of proinflammatory cytokine or anti-inflammatory cytokine. However, in addition to the limited scope of the study, a single short duration of CO exposure (1 h) may not have been sufficient to gain the benefits of CO in human subjects. Subsequently, ex-smoking patients with stable chronic obstructive pulmonary disease were subjected to CO inhalation with 100–125 ppm for 2 h/day for 4 days (5). After the exposure CO-Hb was increased to 4.5% in the patients. Importantly, patients with CO inhalation showed decrease number of eosinophils in sputum and improvement of methacholine responsiveness (5). In a recent completed trial of Inhaled Carbon Monoxide to Treat Idiopathic Pulmonary Fibrosis (NCT01214187), inhaled CO at 100–200 ppm was administered two times weekly for 2 h per dose to complete 12 wk of treatment (Table 1). Although the full analysis of this completed trial is pending and additional studies are needed in the future to assess the efficacy of CO on idiopathic pulmonary fibrosis, this trial has importantly demonstrated that inhaled CO at 100–200 ppm can be administered safely to patients with underlying lung dysfunction. Finally, a new clinical trial of CO, Safety Study of Inhaled Carbon Monoxide to Treat Acute Respiratory Distress Syndrome (ARDS) (NCT02425579), has started recruiting participants (Table 1). The purpose of the trial is to assess the safety of inhaled CO in intubated patients with sepsis-induced ARDS. Since sepsis is still one of the leading causes of ARDS, this new clinical trial may provide the critical information how CO can be efficiently and safely delivered to septic patients.

Table 1.

List of ongoing clinical trials using CO inhalation in human diseases

Target Disease Phase Purpose Assigned Interventions Primary Outcome Measures Second Outcome Measures
Acute Respiratory Distress Syndrome (ARDS) (NCT02425579) 1
  • To assess the safety of inhaled CO in intubated patients with sepsis-induced ARDS

  • Inhaled CO at 100 or 200 ppm

  • Algorithm-specified dose of inhaled CO to achieve CO-Hb of 6–8%

  • Number of administration associated adverse events

[time frame: 60 days]
  • Mean daily SOFA score

  • PaO2/FiO2 ratio and oxygenation index

  • ALI score

  • Vasopressor-free days

  • ICU-free days

  • Hospital-free days

  • Plasma biomarkers of inflammation, lung epithelial injury, endothelial injury, markers of change in other end-organ function

  • Skeletal muscle biomarkers of mitochondrial biogenesis

Intestinal Paralysis After Colon Surgery (NCT01050712) 2
  • To determine whether inhaling very low doses of CO before and after colon surgery will shorten the duration of normal POI and/or prevent the development of POI complications in patients undergoing colon surgery

  • Concentration of CO to be inhaled by patients will be determined in a safety trial performed in healthy volunteers prior to the commencement of this trial

  • Duration of POI (radiological)

  • Incidence of pathological POI

  • Duration of POI (clinical)

  • Patients will receive CO for 1 h prior to colon resection and for 1 h after colon resection

Aortic Valve Surgery Patients (NCT01727167) 1 and 2
  • To test whether inhalation of CO will increase the numbers of mitochondria in heart muscle

  • 200 ppm of CO for 1 h on the day prior to surgery

  • Biochemical markers for mitochondrial biogenesis (blood and right atrial tissue)

  • To compare blood to right atrial tissue biochemical markers of mitochondrial biogenesis

  • Right atrial biochemical markers will be measured one time only, intra-operatively

  • Biochemical markers in both right atrial tissue and blood will be measured and compared to see whether the more easily obtained blood markers accurately describe changes expected in the heart

  • Blood biochemical markers will be measured before CO exposure and at intervals up to one week postoperatively

Pulmonary Arterial Hypertension (NCT01523548) 1 and 2
  • To examine the potential of CO to decrease elevated blood pressure in the pulmonary artery

  • 150 ppm × 3 h once weekly (week 1) 150 ppm × 3 h twice weekly (week 2) ·150 ppm × 3 h three times a week (weeks 3–16)

  • Evidence of a 20% decrease in pulmonary vascular resistance posttherapy when compared to pretherapy value

  • Effect of 16-wk CO inhalation on other pulmonary and systemic hemodynamic parameters

  • Effect of 16-wk CO inhalation on functional capacity assessed by 6-min walk test

  • Effect of 16-wk CO inhalation on brain natriuretic peptide levels

  • Effect of 16-wk CO inhalation on right ventricular echocardiographic parameters

  • Effect of 16-wk CO inhalation on acute pulmonary vasoreactivity

Idiopathic Pulmonary Fibrosis (NCT01214187) 2
  • To determine whether low-concentration inhaled carbon monoxide is effective in treating idiopathic pulmonary fibrosis

  • The intervention will be inhaled CO at 100–200 ppm administered two times weekly for 2 h per dose to complete 12 wk of treatment

  • Serum MMP7 level

  • Total lung capacity

[time frame: 3 mo]
  • Diffusing capacity for CO

  • 6-min walk distance

  • St. George's Respiratory Questionnaire

ALI, acute lung injury; ARDS, acute respiratory distress syndrome; FiO2, inspired O2 fraction; ICU, intensive care unit; PaO2, arterial partial pressure of O2; POI, postoperative ileus; SOFA, sequential organ failure assessment. Detailed information is available at https://clinicaltrials.gov/.

The Future of CO Therapy in Sepsis

The first report of protective roles of inhaled CO was demonstrated in rodents with hyperoxia-induced ALI in 1999 (49). Low-dose CO exposure (250 ppm) reduced the volume of pleural effusion and the protein concentration and neutrophil recruitment in the airways (49). Furthermore, this study also demonstrated that CO inhalation alone is enough to protect lung injuries regardless of endogenous CO production in the rodent models (49). In the past 15 years, CO inhalation has been successfully shown as a potential therapeutic in a number of animal models, and the inhalation of CO gas has now been tested in multiple early phase I clinical trials (Table 1). Although beneficial roles of low-dose CO inhalation are proven in rodent models, the studies of inhaled CO as the treatment for human diseases are at an early phase in the clinical development. Further studies are needed to establish the optimal delivery and dosing protocol for CO inhalation therapy in critically ill patients (e.g., differences in lung physiology and mechanics between rodents and humans should be kept in mind). As much as we are most committed and excited by the potential therapeutic applicability of CO in sepsis going forward, we must remain vigilant in designing the optimal treatment protocol and further works still lies ahead. Patients who have risk of neurotoxicity or carboxyhemoglobinemia may be excluded from CO inhalation therapy (the possible exclusion criteria are listed on https://clinicaltrials.gov/ct2/show/NCT02425579). Nonetheless, given a dreadful disease state such as sepsis with no effective therapeutic target and enormous unmet medical need, the potent protective molecule CO is hard to be overlooked but should be evaluated for the benefits of patients. We have entered an exciting era wherein Phase I CO trials have been completed in several disease states: there are challenges ahead of us to ensure that through carefully planned Phase II trials we can rigorously test the tenet whether CO inhalation therapy will be effectively tested in clinical trials and in the future can represent a viable treatment among the available therapeutic armamentariums for patients suffering from sepsis.

GRANTS

This work was funded by National Heart, Lung, and Blood Institute Grants P01 HL1008801 and R01 HL079904 to A. M. K. Choi.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the author(s).

AUTHOR CONTRIBUTIONS

K.N. and A.M.C. conception and design of research; K.N. prepared figures; K.N. drafted manuscript; K.N. and A.M.C. edited and revised manuscript; A.M.C. approved final version of manuscript.

REFERENCES

  • 1.Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med 369: 2063, 2013. [DOI] [PubMed] [Google Scholar]
  • 2.Annane D, Timsit JF, Megarbane B, Martin C, Misset B, Mourvillier B, Siami S, Chagnon JL, Constantin JM, Petitpas F, Souweine B, Amathieu R, Forceville X, Charpentier C, Tesniere A, Chastre J, Bohe J, Colin G, Cariou A, Renault A, Brun-Buisson C, Bellissant E; APROCCHSS Trial Investigators. Recombinant human activated protein C for adults with septic shock: a randomized controlled trial. Am J Respir Crit Care Med 187: 1091–1097, 2013. [DOI] [PubMed] [Google Scholar]
  • 3.Antuni JD, Kharitonov SA, Hughes D, Hodson ME, Barnes PJ. Increase in exhaled carbon monoxide during exacerbations of cystic fibrosis. Thorax 55: 138–142, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Anyanwu AC, Bentley JK, Popova AP, Malas O, Alghanem H, Goldsmith AM, Hershenson MB, Pinsky DJ. Suppression of inflammatory cell trafficking and alveolar simplification by the heme oxygenase-1 product carbon monoxide. Am J Physiol Lung Cell Mol Physiol 306: L749–L763, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bathoorn E, Slebos DJ, Postma DS, Koeter GH, van Oosterhout AJ, van der Toorn M, Boezen HM, Kerstjens HA. Anti-inflammatory effects of inhaled carbon monoxide in patients with COPD: a pilot study. Eur Respir J 30: 1131–1137, 2007. [DOI] [PubMed] [Google Scholar]
  • 6.Brealey D, Singer M. Mitochondrial dysfunction in sepsis. Curr Infect Dis Rep 5: 365–371, 2003. [DOI] [PubMed] [Google Scholar]
  • 7.Chiang N, Fredman G, Backhed F, Oh SF, Vickery T, Schmidt BA, Serhan CN. Infection regulates pro-resolving mediators that lower antibiotic requirements. Nature 484: 524–528, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Chiang N, Shinohara M, Dalli J, Mirakaj V, Kibi M, Choi AM, Serhan CN. Inhaled carbon monoxide accelerates resolution of inflammation via unique proresolving mediator-heme oxygenase-1 circuits. J Immunol 190: 6378–6388, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chung SW, Liu X, Macias AA, Baron RM, Perrella MA. Heme oxygenase-1-derived carbon monoxide enhances the host defense response to microbial sepsis in mice. J Clin Invest 118: 239–247, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Coburn RF. The carbon monoxide body stores. Ann NY Acad Sci 174: 11–22, 1970. [DOI] [PubMed] [Google Scholar]
  • 11.Csongradi E, Juncos LA, Drummond HA, Vera T, Stec DE. Role of carbon monoxide in kidney function: is a little carbon monoxide good for the kidney? Curr Pharm Biotechnol 13: 819–826, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dalli J, Kraft BD, Colas RA, Shinohara M, Fredenburgh LE, Hess DR, Chiang N, Welty-Wolf K, Choi AM, Piantadosi CA, Serhan CN. The regulation of proresolving lipid mediator profiles in baboon pneumonia by inhaled carbon monoxide. Am J Respir Cell Mol Biol 53: 314–325, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Desmard M, Davidge KS, Bouvet O, Morin D, Roux D, Foresti R, Ricard JD, Denamur E, Poole RK, Montravers P, Motterlini R, Boczkowski J. A carbon monoxide-releasing molecule (CORM-3) exerts bactericidal activity against Pseudomonas aeruginosa and improves survival in an animal model of bacteraemia. FASEB J 23: 1023–1031, 2009. [DOI] [PubMed] [Google Scholar]
  • 14.Dolinay T, Kim YS, Howrylak J, Hunninghake GM, An CH, Fredenburgh L, Massaro AF, Rogers A, Gazourian L, Nakahira K, Haspel JA, Landazury R, Eppanapally S, Christie JD, Meyer NJ, Ware LB, Christiani DC, Ryter SW, Baron RM, Choi AM. Inflammasome-regulated cytokines are critical mediators of acute lung injury. Am J Respir Crit Care Med 185: 1225–1234, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Fredenburgh LE, Kraft BD, Hess DR, Harris RS, Wolf MA, Suliman HB, Roggli VL, Davies JD, Winkler T, Stenzler A, Baron RM, Thompson BT, Choi AM, Welty-Wolf KE, Piantadosi CA. Effects of inhaled CO administration on acute lung injury in baboons with pneumococcal pneumonia. Am J Physiol Lung Cell Mol Physiol 309: L834–L846, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gentile MA. Inhaled medical gases: more to breathe than oxygen. Respir Care 56: 1341–1357; discussion 1357–1349, 2011. [DOI] [PubMed] [Google Scholar]
  • 17.Gorman D, Drewry A, Huang YL, Sames C. The clinical toxicology of carbon monoxide. Toxicology 187: 25–38, 2003. [DOI] [PubMed] [Google Scholar]
  • 18.Gozzelino R, Jeney V, Soares MP. Mechanisms of cell protection by heme oxygenase-1. Annu Rev Pharmacol Toxicol 50: 323–354, 2010. [DOI] [PubMed] [Google Scholar]
  • 19.Grundler K, Angstwurm M, Hilge R, Baumann P, Annecke T, Crispin A, Sohn HY, Massberg S, Kraemer BF. Platelet mitochondrial membrane depolarization reflects disease severity in patients with sepsis and correlates with clinical outcome. Crit Care 18: R31, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med 21: 677–687, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hall MJ, Williams SN, DeFrances CJ, Golosinskiy A. Inpatient care for septicemia or sepsis: a challenge for patients and hospitals. NCHS Data Brief 1–8, 2011. [PubMed] [Google Scholar]
  • 22.Horvath I, Loukides S, Wodehouse T, Kharitonov SA, Cole PJ, Barnes PJ. Increased levels of exhaled carbon monoxide in bronchiectasis: a new marker of oxidative stress. Thorax 53: 867–870, 1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ignarro LJ, Buga GM, Wood KS, Byrns RE, Chaudhuri G. Endothelium-derived relaxing factor produced and released from artery and vein is nitric oxide. Proc Natl Acad Sci USA 84: 9265–9269, 1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ignarro LJ, Byrns RE, Buga GM, Wood KS. Endothelium-derived relaxing factor from pulmonary artery and vein possesses pharmacologic and chemical properties identical to those of nitric oxide radical. Circ Res 61: 866–879, 1987. [DOI] [PubMed] [Google Scholar]
  • 25.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 308: L1058–L1067, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kim SI, Na HJ, Ding Y, Wang Z, Lee SJ, Choi ME. Autophagy promotes intracellular degradation of type I collagen induced by transforming growth factor (TGF)-beta1. J Biol Chem 287: 11677–11688, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Krysko DV, Agostinis P, Krysko O, Garg AD, Bachert C, Lambrecht BN, Vandenabeele P. Emerging role of damage-associated molecular patterns derived from mitochondria in inflammation. Trends Immunol 32: 157–164, 2011. [DOI] [PubMed] [Google Scholar]
  • 28.Kung CT, Hsiao SY, Tsai TC, Su CM, Chang WN, Huang CR, Wang HC, Lin WC, Chang HW, Lin YJ, Cheng BC, Su BY, Tsai NW, Lu CH. Plasma nuclear and mitochondrial DNA levels as predictors of outcome in severe sepsis patients in the emergency room. J Transl Med 10: 130, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell 157: 1013–1022, 2014. [DOI] [PubMed] [Google Scholar]
  • 30.Lee S, Lee SJ, Coronata AA, Fredenburgh LE, Chung SW, Perrella MA, Nakahira K, Ryter SW, Choi AM. Carbon monoxide confers protection in sepsis by enhancing beclin 1-dependent autophagy and phagocytosis. Antioxid Redox Signal 20: 432–442, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lee SJ, Ryter SW, Xu JF, Nakahira K, Kim HP, Choi AM, Kim YS. Carbon monoxide activates autophagy via mitochondrial reactive oxygen species formation. Am J Respir Cell Mol Biol 45: 867–873, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Levy BD, Serhan CN. Resolution of acute inflammation in the lung. Annu Rev Physiol 76: 467–492, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Liu QP, Zhou DX, Lin P, Gao XL, Pan L, Jin FG. Participation of autophagy in acute lung injury induced by seawater. Exp Lung Res 39: 441–452, 2013. [DOI] [PubMed] [Google Scholar]
  • 34.Liu SH, Xu XR, Ma K, Xu B. Protection of carbon monoxide inhalation on lipopolysaccharide-induced multiple organ injury in rats. Chin Med Sci J 22: 169–176, 2007. [PubMed] [Google Scholar]
  • 35.Liu V, Escobar GJ, Greene JD, Soule J, Whippy A, Angus DC, Iwashyna TJ. Hospital deaths in patients with sepsis from 2 independent cohorts. JAMA 312: 90–92, 2014. [DOI] [PubMed] [Google Scholar]
  • 36.Marshall JC. Why have clinical trials in sepsis failed? Trends Mol Med 20: 195–203, 2014. [DOI] [PubMed] [Google Scholar]
  • 37.Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med 348: 1546–1554, 2003. [DOI] [PubMed] [Google Scholar]
  • 38.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 171: 354–360, 2005. [DOI] [PubMed] [Google Scholar]
  • 39.Mazzola S, Forni M, Albertini M, Bacci ML, Zannoni A, Gentilini F, Lavitrano M, Bach FH, Otterbein LE, Clement MG. Carbon monoxide pretreatment prevents respiratory derangement and ameliorates hyperacute endotoxic shock in pigs. FASEB J 19: 2045–2047, 2005. [DOI] [PubMed] [Google Scholar]
  • 40.Moncada S, Palmer RM, Higgs EA. Nitric oxide: physiology, pathophysiology, pharmacology. Pharmacol Rev 43: 109–142, 1991. [PubMed] [Google Scholar]
  • 41.Moore BA, Otterbein LE, Turler A, Choi AM, Bauer AJ. Inhaled carbon monoxide suppresses the development of postoperative ileus in the murine small intestine. Gastroenterology 124: 377–391, 2003. [DOI] [PubMed] [Google Scholar]
  • 42.Morse D, Pischke SE, Zhou Z, Davis RJ, Flavell RA, Loop T, Otterbein SL, Otterbein LE, Choi AM. Suppression of inflammatory cytokine production by carbon monoxide involves the JNK pathway and AP-1. J Biol Chem 278: 36993–36998, 2003. [DOI] [PubMed] [Google Scholar]
  • 43.Motterlini R, Otterbein LE. The therapeutic potential of carbon monoxide. Nat Rev Drug Discov 9: 728–743, 2010. [DOI] [PubMed] [Google Scholar]
  • 44.Nakahira K, Choi AM. Autophagy: a potential therapeutic target in lung diseases. Am J Physiol Lung Cell Mol Physiol 305: L93–L107, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Nakahira K, Hisata S, Choi AM. The roles of mitochondrial damage-associated molecular patterns in diseases. Antioxid Redox Signaling 2015. Aug 17. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.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 203: 2377–2389, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Nakahira K, Kyung SY, Rogers AJ, Gazourian L, Youn S, Massaro AF, Quintana C, Osorio JC, Wang Z, Zhao Y, Lawler LA, Christie JD, Meyer NJ, Mc Causland FR, Waikar SS, Waxman AB, Chung RT, Bueno R, Rosas IO, Fredenburgh LE, Baron RM, Christiani DC, Hunninghake GM, Choi AM. Circulating mitochondrial DNA in patients in the ICU as a marker of mortality: derivation and validation. PLoS Med 10: e1001577; discussion e1001577, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Otterbein LE, Bach FH, Alam J, Soares M, Tao Lu H, Wysk M, Davis RJ, Flavell RA, Choi AM. Carbon monoxide has anti-inflammatory effects involving the mitogen-activated protein kinase pathway. Nat Med 6: 422–428, 2000. [DOI] [PubMed] [Google Scholar]
  • 49.Otterbein LE, Mantell LL, Choi AM. Carbon monoxide provides protection against hyperoxic lung injury. Am J Physiol Lung Cell Mol Physiol 276: L688–L694, 1999. [DOI] [PubMed] [Google Scholar]
  • 50.Otterbein LE, May A, Chin BY. Carbon monoxide increases macrophage bacterial clearance through Toll-like receptor (TLR)4 expression. Cell Mol Biol 51: 433–440, 2005. [PubMed] [Google Scholar]
  • 51.Paine A, Eiz-Vesper B, Blasczyk R, Immenschuh S. Signaling to heme oxygenase-1 and its anti-inflammatory therapeutic potential. Biochem Pharmacol 80: 1895–1903, 2010. [DOI] [PubMed] [Google Scholar]
  • 52.Piantadosi CA. Diagnosis and treatment of carbon monoxide poisoning. Respir Care Clin N Am 5: 183–202, 1999. [PubMed] [Google Scholar]
  • 53.Ranieri VM, Thompson BT, Barie PS, Dhainaut JF, Douglas IS, Finfer S, Gardlund B, Marshall JC, Rhodes A, Artigas A, Payen D, Tenhunen J, Al-Khalidi HR, Thompson V, Janes J, Macias WL, Vangerow B, Williams MD; PROWESS-SHOCK Study Group. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med 366: 2055–2064, 2012. [DOI] [PubMed] [Google Scholar]
  • 54.Russell JA. Management of sepsis. N Engl J Med 355: 1699–1713, 2006. [DOI] [PubMed] [Google Scholar]
  • 55.Ryter SW, Alam J, Choi AM. Heme oxygenase-1/carbon monoxide: from basic science to therapeutic applications. Physiol Rev 86: 583–650, 2006. [DOI] [PubMed] [Google Scholar]
  • 56.Ryter SW, Choi AM. Carbon monoxide in exhaled breath testing and therapeutics. J Breath Res 7: 017111, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ryter SW, Choi AM. Carbon monoxide: present and future indications for a medical gas. Korean J Intern Med 28: 123–140, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ryter SW, Choi AM. Targeting heme oxygenase-1 and carbon monoxide for therapeutic modulation of inflammation. Transl Res pii: S1931-5244(15)00216-9, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ryter SW, Kim HP, Nakahira K, Zuckerbraun BS, Morse D, Choi AM. Protective functions of heme oxygenase-1 and carbon monoxide in the respiratory system. Antioxid Redox Signal 9: 2157–2173, 2007. [DOI] [PubMed] [Google Scholar]
  • 60.Ryter SW, Morse D, Choi AM. Carbon monoxide: to boldly go where NO has gone before. Sci STKE 2004: RE6, 2004. [DOI] [PubMed] [Google Scholar]
  • 61.Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature 510: 92–101, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Serhan CN, Chiang N, Dalli J. The resolution code of acute inflammation: novel pro-resolving lipid mediators in resolution. Semin Immunol 27: 200–215, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Shinohara M, Kibi M, Riley IR, Chiang N, Dalli J, Kraft BD, Piantadosi CA, Choi AM, Serhan CN. Cell-cell interactions and bronchoconstrictor eicosanoid reduction with inhaled carbon monoxide and resolvin D1. Am J Physiol Lung Cell Mol Physiol 307: L746–L757, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Singer M. The role of mitochondrial dysfunction in sepsis-induced multi-organ failure. Virulence 5: 66–72, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Siriussawakul A, Chen LI, Lang JD. Medical gases: a novel strategy for attenuating ischemia-reperfusion injury in organ transplantation? J Transplant 2012: 819382, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Song R, Zhou Z, Kim PK, Shapiro RA, Liu F, Ferran C, Choi AM, Otterbein LE. Carbon monoxide promotes Fas/CD95-induced apoptosis in Jurkat cells. J Biol Chem 279: 44327–44334, 2004. [DOI] [PubMed] [Google Scholar]
  • 67.Spite M, Norling LV, Summers L, Yang R, Cooper D, Petasis NA, Flower RJ, Perretti M, Serhan CN. Resolvin D2 is a potent regulator of leukocytes and controls microbial sepsis. Nature 461: 1287–1291, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Stearns-Kurosawa DJ, Osuchowski MF, Valentine C, Kurosawa S, Remick DG. The pathogenesis of sepsis. Annu Rev Pathol 6: 19–48, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sun Y, Li C, Shu Y, Ju X, Zou Z, Wang H, Rao S, Guo F, Liu H, Nan W, Zhao Y, Yan Y, Tang J, Zhao C, Yang P, Liu K, Wang S, Lu H, Li X, Tan L, Gao R, Song J, Gao X, Tian X, Qin Y, Xu KF, Li D, Jin N, Jiang C. Inhibition of autophagy ameliorates acute lung injury caused by avian influenza A H5N1 infection. Sci Signal 5: ra16, 2012. [DOI] [PubMed] [Google Scholar]
  • 70.Svistunenko DA, Davies N, Brealey D, Singer M, Cooper CE. Mitochondrial dysfunction in patients with severe sepsis: an EPR interrogation of individual respiratory chain components. Biochim Biophys Acta 1757: 262–272, 2006. [DOI] [PubMed] [Google Scholar]
  • 71.Takagi T, Naito Y, Uchiyama K, Yoshikawa T. The role of heme oxygenase and carbon monoxide in inflammatory bowel disease. Redox Rep 15: 193–201, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Von Burg R. Carbon monoxide. J Appl Toxicol 19: 379–386, 1999. [DOI] [PubMed] [Google Scholar]
  • 73.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 282: 1718–1726, 2007. [DOI] [PubMed] [Google Scholar]
  • 74.Wegiel B, Larsen R, Gallo D, Chin BY, Harris C, Mannam P, Kaczmarek E, Lee PJ, Zuckerbraun BS, Flavell R, Soares MP, Otterbein LE. Macrophages sense and kill bacteria through carbon monoxide-dependent inflammasome activation. J Clin Invest 124: 4926–4940, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Weiss SL, Selak MA, Tuluc F, Perales Villarroel J, Nadkarni VM, Deutschman CS, Becker LB. Mitochondrial dysfunction in peripheral blood mononuclear cells in pediatric septic shock. Pediatr Crit Care Med 16: e4–e12, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Whiteman M, Le Trionnaire S, Chopra M, Fox B, Whatmore J. Emerging role of hydrogen sulfide in health and disease: critical appraisal of biomarkers and pharmacological tools. Clin Sci (Lond) 121: 459–488, 2011. [DOI] [PubMed] [Google Scholar]
  • 77.Williams SC. After Xigris, researchers look to new targets to combat sepsis. Nat Med 18: 1001, 2012. [DOI] [PubMed] [Google Scholar]
  • 78.Yamakawa K, Ogura H, Koh T, Ogawa Y, Matsumoto N, Kuwagata Y, Shimazu T. Platelet mitochondrial membrane potential correlates with severity in patients with systemic inflammatory response syndrome. J Trauma Acute Care Surg 74: 411–417; discussion 418, 2013. [DOI] [PubMed] [Google Scholar]
  • 79.Zegdi R, Perrin D, Burdin M, Boiteau R, Tenaillon A. Increased endogenous carbon monoxide production in severe sepsis. Intensive Care Med 28: 793–796, 2002. [DOI] [PubMed] [Google Scholar]
  • 80.Zhang J, Yao X, Yu R, Bai J, Sun Y, Huang M, Adcock IM, Barnes PJ. Exhaled carbon monoxide in asthmatics: a meta-analysis. Respir Res 11: 50, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zuckerbraun BS, Billiar TR, Otterbein SL, Kim PK, Liu F, Choi AM, Bach FH, Otterbein LE. Carbon monoxide protects against liver failure through nitric oxide-induced heme oxygenase 1. J Exp Med 198: 1707–1716, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Lung Cellular and Molecular Physiology are provided here courtesy of American Physiological Society

RESOURCES