T. Li, H. L. Bonkovsky, and J. T. Guo, Structural analysis of heme proteins: implications for design and prediction, BMC Struct Biol, vol.11, p.13, 2011.

M. D. Maines, Overview of heme degradation pathway, Curr Protoc Toxicol, 2001.

S. W. Ryter and R. M. Tyrrell, The heme synthesis and degradation pathways: role in oxidant sensitivity. Heme oxygenase has both pro-and antioxidant properties, Free Radic Biol Med, vol.28, pp.289-309, 2000.

P. A. Dennery, Signaling function of heme oxygenase proteins, Antioxid Redox Signal, vol.20, pp.1743-53, 2014.

M. D. Maines, Heme oxygenase: function, multiplicity, regulatory mechanisms, and clinical applications, FASEB J, vol.2, pp.2557-68, 1988.

N. G. Abraham, J. Cao, D. Sacerdoti, X. Li, and G. Drummond, Heme oxygenase: the key to renal function regulation, Am J Physiol Renal Physiol, vol.297, pp.1137-52, 2009.

G. Kikuchi, T. Yoshida, and M. Noguchi, Heme oxygenase and heme degradation, Biochem Biophys Res Commun, vol.338, pp.558-67, 2005.

M. Bauer and I. Bauer, Heme oxygenase-1: redox regulation and role in the hepatic response to oxidative stress, Antioxid Redox Signal, vol.4, pp.749-58, 2002.

Y. Zhang, G. Jiang, M. Sauler, and P. J. Lee, Lung endothelial HO-1 targeting in vivo using lentiviral miRNA regulates apoptosis and autophagy during oxidant injury, FASEB J, vol.27, pp.4041-58, 2013.

D. Morse, S. E. Pischke, Z. Zhou, R. J. Davis, R. A. Flavell et al., Suppression of inflammatory cytokine production by carbon monoxide involves the JNK pathway and AP-1, J Biol Chem, vol.278, pp.36993-37001, 2003.

C. Chauveau, S. Remy, P. J. Royer, M. Hill, S. Tanguy-royer et al., Heme oxygenase-1 expression inhibits dendritic cell maturation and proinflammatory function but conserves IL-10 expression, Blood, vol.106, pp.1694-702, 2005.

D. Morse, The role of heme oxygenase-1 in pulmonary fibrosis, Am J Respir Cell Mol Biol, vol.29, pp.82-88, 2003.

M. D. Maines, G. M. Trakshel, and R. K. Kutty, Characterization of two constitutive forms of rat liver microsomal heme oxygenase. Only one molecular species of the enzyme is inducible, J Biol Chem, vol.261, pp.411-420, 1986.

G. M. Trakshel, R. K. Kutty, and M. D. Maines, Purification and characterization of the major constitutive form of testicular heme oxygenase. The noninducible isoform, J Biol Chem, vol.261, pp.11131-11138, 1986.

J. M. Stout, M. U. Gousset, H. A. Drummond, . Gray-w-3rd, B. E. Pruett et al., Sex-specific effects of heme oxygenase-2 deficiency on renovascular hypertension, J Am Soc Hypertens, vol.7, pp.328-363, 2013.

S. Hayashi, Y. Omata, H. Sakamoto, Y. Higashimoto, T. Hara et al., Characterization of rat heme oxygenase-3 gene. Implication of processed pseudogenes derived from heme oxygenase-2 gene, Gene, vol.336, pp.241-50, 2004.

N. Radhakrishnan, S. P. Yadav, A. Sachdeva, P. K. Pruthi, S. Sawhney et al., Human heme oxygenase-1 deficiency presenting with hemolysis, nephritis, and asplenia, J Pediatr Hematol Oncol, vol.33, pp.74-82, 2011.

K. D. Poss and S. Tonegawa, Reduced stress defense in heme oxygenase 1-deficient cells, Proc Natl Acad Sci, vol.94, pp.10925-10955, 1997.

A. Yachie, Y. Niida, T. Wada, N. Igarashi, H. Kaneda et al., Oxidative stress causes enhanced endothelial cell injury in human heme oxygenase-1 deficiency, J Clin Invest, vol.103, pp.129-164, 1999.

J. P. Mackern-oberti, S. A. Riquelme, C. Llanos, C. B. Schmidt, T. Simon et al.,

, Heme oxygenase-1 as a target for the design of gene and pharmaceutical therapies for autoimmune diseases, Curr Gene Ther, vol.14, pp.218-253, 2014.

M. H. Kapturczak, C. Wasserfall, T. Brusko, M. Campbell-thompson, T. M. Ellis et al., Heme oxygenase-1 modulates early inflammatory responses: evidence from the heme oxygenase-1-deficient mouse, Am J Pathol, vol.165, pp.1045-53, 2004.

A. A. Herrada, C. Llanos, J. P. Mackern-oberti, L. J. Carreno, C. Henriquez et al., Haem oxygenase 1 expression is altered in monocytes from patients with systemic lupus erythematosus, Immunology, vol.136, pp.414-438, 2012.

P. Fagone, F. Patti, K. Mangano, S. Mammana, M. Coco et al., Heme oxygenase-1 expression in peripheral blood mononuclear cells correlates with disease activity in multiple sclerosis, J Neuroimmunol, vol.261, pp.82-88, 2013.

E. Y. Ryu, S. Y. Park, S. G. Kim, J. Da-park, J. S. Kang et al., Anti-inflammatory effect of heme oxygenase-1 toward Porphyromonas gingivalis lipopolysaccharide in macrophages exposed to gomisins A, G, and J, J Med Food, vol.14, pp.1519-1545, 2011.

T. D. Hull, A. Agarwal, and J. F. George, The mononuclear phagocyte system in homeostasis and disease: a role for heme oxygenase-1, Antioxid Redox Signal, vol.20, pp.1770-88, 2014.

H. G. Chen, K. L. Xie, H. Z. Han, W. N. Wang, D. Q. Liu et al., Heme oxygenase-1 mediates the anti-inflammatory effect of molecular hydrogen in LPS-stimulated RAW 264.7 macrophages, Int J Surg, vol.11, pp.1060-1066, 2013.

S. Remy, P. Blancou, L. Tesson, V. Tardif, R. Brion et al., Carbon monoxide inhibits TLR-induced dendritic cell immunogenicity, J Immunol, vol.182, pp.1877-84, 2009.

D. De-nardo, Toll-like receptors: activation, signalling and transcriptional modulation, Cytokine, vol.74, pp.181-190, 2015.

B. Wegiel, C. J. Hauser, and L. E. Otterbein, Heme as a danger molecule in pathogen recognition, Free Radic Biol Med, vol.89, pp.651-61, 2015.

M. Cargnello and P. P. Roux, Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases, Microbiol Mol Biol Rev, vol.75, pp.50-83, 2011.

C. Davies and C. Tournier, Exploring the function of the JNK (c-Jun N-terminal kinase) signalling pathway in physiological and pathological processes to design novel therapeutic strategies, Biochem Soc Trans, vol.40, pp.85-94, 2012.

S. A. Rushworth, X. Chen, N. Mackman, R. M. Ogborne, O. Connell et al., Lipopolysaccharide-induced heme oxygenase-1 expression in human monocytic cells is mediated via Nrf2 and protein kinase C, J Immunol, vol.175, pp.4408-4423, 2005.

K. Srisook and Y. N. Cha, Super-induction of HO-1 in macrophages stimulated with lipopolysaccharide by prior depletion of glutathione decreases iNOS expression and NO production, Nitric Oxide, vol.12, pp.70-79, 2005.

H. C. Huang, T. Nguyen, and C. B. Pickett, Phosphorylation of Nrf2 at Ser-40 by protein kinase C regulates antioxidant response element-mediated transcription, J Biol Chem, vol.277, pp.42769-74, 2002.

T. S. Lee and L. Y. Chau, Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice, Nat Med, vol.8, pp.240-246, 2002.

L. M. Al-huseini, A. Yeang, H. X. Sethu, S. Alhumeed, N. Hamdam et al., Nuclear factor-erythroid 2 (NF-E2) p45-related factor-2 (Nrf2) modulates dendritic cell immune function through regulation of p38 MAPK-cAMP-responsive element binding protein/activating transcription factor 1 signaling, J Biol Chem, vol.288, pp.22281-22289, 2013.

K. Igarashi and J. Sun, The heme-Bach1 pathway in the regulation of oxidative stress response and erythroid differentiation, Antioxid Redox Signal, vol.8, pp.107-125, 2006.

S. Jang, J. Piao, S. Cha, Y. N. Kim, and C. , Taurine chloramine activates Nrf2, increases HO-1 expression and protects cells from death caused by hydrogen peroxide, J Clin Biochem Nutr, vol.45, pp.37-43, 2009.

K. H. Pulkkinen, S. Yla-herttuala, and A. L. Levonen, Heme oxygenase 1 is induced by miR-155 via reduced BACH1 translation in endothelial cells, Free Radic Biol Med, vol.51, pp.2124-2155, 2011.

C. E. Mccoy, F. J. Sheedy, J. E. Qualls, S. L. Doyle, S. R. Quinn et al., IL-10 inhibits miR-155 induction by toll-like receptors, J Biol Chem, vol.285, pp.20492-20500, 2010.

Q. Ma, L. Battelli, and A. F. Hubbs, Multiorgan autoimmune inflammation, enhanced lymphoproliferation, and impaired homeostasis of reactive oxygen species in mice lacking the antioxidant-activated transcription factor Nrf2, Am J Pathol, vol.168, pp.1960-74, 2006.

M. Exner, E. Minar, O. Wagner, and M. Schillinger, The role of heme oxygenase-1 promoter polymorphisms in human disease, Free Radic Biol Med, vol.37, pp.1097-104, 2004.

B. Rueda, J. Oliver, G. Robledo, M. A. Lopez-nevot, A. Balsa et al., HO-1 promoter polymorphism associated with rheumatoid arthritis, Arthritis Rheum, vol.56, pp.3953-3961, 2007.

N. Yamada, M. Yamaya, S. Okinaga, K. Nakayama, K. Sekizawa et al., Microsatellite polymorphism in the heme oxygenase-1 gene promoter is associated with susceptibility to emphysema, Am J Hum Genet, vol.66, pp.187-95, 2000.

C. Sponholz, K. Huse, M. Kramer, E. J. Giamarellos-bourboulis, R. A. Claus et al., Gene polymorphisms in the heme degradation pathway and outcome of severe human sepsis, Shock, vol.38, pp.459-65, 2012.

K. Ono, T. Mannami, and N. Iwai, Association of a promoter variant of the haeme oxygenase-1 gene with hypertension in women, J Hypertens, vol.21, pp.1497-503, 2003.

K. Ono, Y. Goto, S. Takagi, S. Baba, N. Tago et al., A promoter variant of the heme oxygenase-1 gene may reduce the incidence of ischemic heart disease in Japanese, Atherosclerosis, vol.173, pp.315-324, 2004.

D. J. Askenazi, B. Halloran, N. Patil, S. Keeling, B. Saeidi et al., Genetic polymorphisms of heme-oxygenase 1 (HO-1) may impact on acute kidney injury, bronchopulmonary dysplasia, and mortality in premature infants, Pediatr Res, vol.77, pp.793-801, 2015.

M. Hajdena-dawson, W. Zhang, P. R. Contag, R. J. Wong, H. J. Vreman et al., Effects of metalloporphyrins on heme oxygenase-1 transcription: correlative cell culture assays guide in vivo imaging, Mol Imaging, vol.2, pp.138-187, 2003.

Y. Shan, R. W. Lambrecht, S. E. Donohue, and H. L. Bonkovsky, Role of Bach1 and Nrf2 in upregulation of the heme oxygenase-1 gene by cobalt protoporphyrin, Immunology, vol.20, pp.1-12, 2006.

M. K. Sardana and A. Kappas, Dual control mechanism for heme oxygenase: tin(IV)-protoporphyrin potently inhibits enzyme activity while markedly increasing content of enzyme protein in liver, Proc Natl Acad Sci, vol.84, pp.2464-2472, 1987.

K. E. Anderson, C. S. Simionatto, G. S. Drummond, and A. Kappas, Tissue distribution and disposition of tin-protoporphyrin, a potent competitive inhibitor of heme oxygenase, J Pharmacol Exp Ther, vol.228, pp.327-360, 1984.

C. C. Hung, X. Liu, M. Y. Kwon, Y. H. Kang, S. W. Chung et al., Regulation of heme oxygenase-1 gene by peptidoglycan involves the interaction of Elk-1 and C/EBPa to increase expression, Am J Physiol Lung Cell Mol Physiol, vol.298, pp.870-879, 2010.

S. Akira and K. Takeda, Toll-like receptor signalling, Nat Rev Immunol, vol.4, pp.499-511, 2004.

S. Akira, M. Yamamoto, and K. Takeda, Role of adapters in Toll-like receptor signalling, Biochem Soc Trans, vol.31, pp.637-679, 2003.

L. A. O'neill, D. Golenbock, and A. G. Bowie, The history of Toll-like receptors -redefining innate immunity, Nat Rev Immunol, vol.13, pp.453-60, 2013.

D. A. Ferenbach, V. Ramdas, N. Spencer, L. Marson, I. Anegon et al., Macrophages expressing heme oxygenase-1 improve renal function in ischemia/reperfusion injury, Mol Ther, vol.18, pp.1706-1719, 2010.

J. P. Roach, E. E. Moore, D. A. Partrick, S. S. Damle, C. C. Silliman et al., Heme oxygenase-1 induction in macrophages by a hemoglobin-based oxygen carrier reduces endotoxin-stimulated cytokine secretion, Shock, vol.31, pp.251-258, 2009.

C. Chauveau, D. Bouchet, J. C. Roussel, P. Mathieu, C. Braudeau et al., Gene transfer of heme oxygenase-1 and carbon monoxide delivery inhibit chronic rejection, Am J Transplant, vol.2, pp.581-92, 2002.

S. Tzima, P. Victoratos, K. Kranidioti, M. Alexiou, and G. Kollias, Myeloid heme oxygenase-1 regulates innate immunity and autoimmunity by modulating IFN-b production, J Exp Med, vol.206, pp.1167-79, 2009.

T. Laumonier, S. Yang, S. Konig, C. Chauveau, I. Anegon et al., Lentivirus mediated HO-1 gene transfer enhances myogenic precursor cell survival after autologous transplantation in pig, Mol Ther, vol.16, pp.404-414, 2008.

S. A. Riquelme, S. M. Bueno, and A. M. Kalergis, Carbon monoxide down-modulates TLR4/ MD2 expression on innate immune cells and reduces endotoxic shock susceptibility, Immunology, vol.144, pp.321-353, 2014.

J. Xue and A. Habtezion, Carbon monoxide-based therapy ameliorates acute pancreatitis via TLR4 inhibition, J Clin Invest, vol.124, pp.437-484, 2014.

F. Rocuts, Y. Ma, X. Zhang, W. Gao, Y. Yue et al., Carbon monoxide suppresses membrane expression of TLR4 via myeloid differentiation factor-2 in bTC3 cells, J Immunol, vol.185, pp.2134-2143, 2010.

I. Gomez-hurtado, P. Zapater, P. Bellot, S. Pascual, M. Perez-mateo et al., Interleukin-10-mediated heme oxygenase 1-induced underlying mechanism in inflammatory down-regulation by norfloxacin in cirrhosis, Hepatology, vol.53, pp.935-979, 2011.

I. Nassour, B. Kautza, M. Rubin, D. Escobar, J. Luciano et al., Carbon monoxide protects against hemorrhagic shock and resuscitation-induced microcirculatory injury and tissue injury, Shock, vol.43, pp.166-71, 2015.

N. Hettiarachchi, M. Dallas, M. Al-owais, H. Griffiths, N. Hooper et al., Heme oxygenase-1 protects against Alzheimer's amyloid-b1-42-induced toxicity via carbon monoxide production, Cell Death Dis, vol.5, p.1569, 2014.

Y. Ruan, L. Wang, Y. Zhao, Y. Yao, S. Chen et al., Carbon monoxide potently prevents ischemia-induced high-mobility group box 1 translocation and release and protects against lethal renal ischemia-reperfusion injury, Kidney Int, vol.86, pp.525-562, 2014.

A. Nakao and Y. Toyoda, Application of carbon monoxide for transplantation, Curr Pharm Biotechnol, vol.13, pp.827-863, 2012.

J. Banchereau and R. M. Steinman, Dendritic cells and the control of immunity, Nature, vol.392, pp.245-52, 1998.

J. Banchereau, F. Briere, C. Caux, J. Davoust, S. Lebecque et al., Immunobiology of dendritic cells, Annu Rev Immunol, vol.18, pp.767-811, 2000.

S. J. Kim and B. Diamond, Modulation of tolerogenic dendritic cells and autoimmunity, Semin Cell Dev Biol, vol.41, pp.49-58, 2014.

R. M. Steinman, D. Hawiger, and M. C. Nussenzweig, Tolerogenic dendritic cells, Annu Rev Immunol, vol.21, pp.685-711, 2003.

J. P. Mackern-oberti, C. Llanos, L. J. Carreno, S. A. Riquelme, S. H. Jacobelli et al., Carbon monoxide exposure improves immune function in lupus-prone mice, Immunology, vol.140, pp.123-155, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-02165559

L. M. Al-huseini, A. Yeang, H. X. Hamdam, J. M. Sethu, S. Alhumeed et al., Heme oxygenase-1 regulates dendritic cell function through modulation of p38 MAPK-CREB/ATF1 signaling, J Biol Chem, vol.289, pp.16442-51, 2014.

J. Listopad, K. Asadullah, C. Sievers, T. Ritter, C. Meisel et al., Heme oxygenase-1 inhibits T cell-dependent skin inflammation and differentiation and function of antigen-presenting cells, Exp Dermatol, vol.16, pp.661-70, 2007.

C. E. Bunse, V. Fortmeier, S. Tischer, E. Zilian, C. Figueiredo et al., Modulation of heme oxygenase-1 by metalloporphyrins increases anti-viral T cell responses, Clin Exp Immunol, vol.179, pp.265-76, 2015.

C. Braudeau, D. Bouchet, L. Tesson, S. Iyer, S. Remy et al., Induction of longterm cardiac allograft survival by heme oxygenase-1 gene transfer, Gene Ther, vol.11, pp.701-711, 2004.

A. A. Chora, P. Fontoura, A. Cunha, T. F. Pais, S. Cardoso et al., Heme oxygenase-1 and carbon monoxide suppress autoimmune neuroinflammation, J Clin Invest, vol.117, pp.438-485, 2007.

V. Tardif, S. A. Riquelme, R. S. Carreno, L. J. Cortes, C. M. Simon et al., Carbon monoxide decreases endosome-lysosome fusion and inhibits soluble antigen presentation by dendritic cells to T cells, Eur J Immunol, vol.43, pp.2832-2876, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-02166083

E. Gagnon, J. J. Bergeron, and M. Desjardins, ER-mediated phagocytosis: myth or reality?, J Leukoc Biol, vol.77, pp.843-848, 2005.

E. Gagnon, S. Duclos, C. Rondeau, E. Chevet, P. H. Cameron et al., Endoplasmic reticulum-mediated phagocytosis is a mechanism of entry into macrophages, Cell, vol.110, pp.119-150, 2002.

P. Guermonprez and S. Amigorena, Pathways for antigen cross presentation, Springer Semin Immunopathol, vol.26, pp.257-71, 2005.

M. Houde, S. Bertholet, E. Gagnon, S. Brunet, G. Goyette et al., Phagosomes are competent organelles for antigen cross-presentation, Nature, vol.425, pp.402-408, 2003.

N. Zemirli and D. Arnoult, Mitochondrial anti-viral immunity, Int J Biochem Cell Biol, vol.44, pp.1473-1479, 2012.

D. Arnoult, F. Soares, I. Tattoli, and S. E. Girardin, Mitochondria in innate immunity, EMBO Rep, vol.12, pp.901-911, 2011.

O. Haller, G. Kochs, and F. Weber, The interferon response circuit: induction and suppression by pathogenic viruses, Virology, vol.344, pp.119-149, 2006.

A. P. West, I. E. Brodsky, C. Rahner, D. K. Woo, H. Erdjument-bromage et al., TLR signalling augments macrophage bactericidal activity through mitochondrial ROS, Nature, vol.472, pp.476-80, 2011.

C. A. Piantadosi and H. B. Suliman, Redox regulation of mitochondrial biogenesis, Free Radic Biol Med, vol.53, pp.2043-53, 2012.
DOI : 10.1016/j.freeradbiomed.2012.09.014

URL : http://europepmc.org/articles/pmc3604744?pdf=render

M. Roy, P. H. Reddy, M. Iijima, and H. Sesaki, Mitochondrial division and fusion in metabolism, Curr Opin Cell Biol, vol.33, pp.111-119, 2015.
DOI : 10.1016/j.ceb.2015.02.001

URL : http://europepmc.org/articles/pmc4380865?pdf=render

D. M. Turnbull and P. Rustin, Genetic and biochemical intricacy shapes mitochondrial cytopathies, Neurobiol Dis, vol.15, pp.23-29, 2015.
DOI : 10.1016/j.nbd.2015.02.003

M. B. Winter, M. A. Herzik, J. Kuriyan, and M. A. Marletta, Tunnels modulate ligand flux in a heme nitric oxide/oxygen binding (H-NOX) domain, Proc Natl Acad Sci, vol.108, pp.881-890, 2011.

C. Weber, D. J. Cole, D. D. O'regan, and M. C. Payne, Renormalization of myoglobin-ligand binding energetics by quantum many-body effects, Proc Natl Acad Sci, vol.111, pp.5790-5795, 2014.
DOI : 10.1073/pnas.1322966111

URL : http://www.pnas.org/content/111/16/5790.full.pdf

M. Desmard, J. Boczkowski, J. Poderoso, and R. Motterlini, Mitochondrial and cellular heme-dependent proteins as targets for the bioactive function of the heme oxygenase/ carbon monoxide system, Antioxid Redox Signal, vol.9, pp.2139-55, 2007.

R. Long, I. Salouage, A. Berdeaux, R. Motterlini, and D. Morin, CORM-3, a water soluble CO-releasing molecule, uncouples mitochondrial respiration via interaction with the phosphate carrier, Biochim Biophys Acta, vol.1837, pp.201-210, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-00920555

A. S. Almeida, C. Figueiredo-pereira, and H. L. Vieira, Carbon monoxide and mitochondriamodulation of cell metabolism, redox response and cell death, Front Physiol, vol.6, p.33, 2015.
DOI : 10.3389/fphys.2015.00033

URL : https://www.frontiersin.org/articles/10.3389/fphys.2015.00033/pdf

K. Maemura, Q. Zheng, T. Wada, M. Ozaki, S. Takao et al., Reactive oxygen species are essential mediators in antigen presentation by Kupffer cells, Immunol Cell Biol, vol.83, pp.336-379, 2005.
DOI : 10.1111/j.1440-1711.2005.01323.x

L. Bonifaz, M. Cervantes-silva, E. Ontiveros-dotor, E. Lopez-villegas, and F. Sanchez-garcia, A role for mitochondria in antigen processing and presentation, Immunology, vol.144, pp.461-71, 2014.

S. A. Riquelme, J. Pogu, I. Anegon, S. M. Bueno, and A. M. Kalergis, Carbon monoxide impairs mitochondria-dependent endosomal maturation and antigen presentation in dendritic cells, Eur J Immunol, vol.45, pp.3269-88, 2015.
DOI : 10.1002/eji.201545671

URL : https://hal.archives-ouvertes.fr/inserm-02148083

D. J. Slebos, S. W. Ryter, M. Van-der-toorn, F. Liu, F. Guo et al., Mitochondrial localization and function of heme oxygenase-1 in cigarette smoke-induced cell death, Am J Respir Cell Mol Biol, vol.36, pp.409-426, 2007.

S. Bindu, C. Pal, S. Dey, M. Goyal, A. Alam et al., Translocation of heme oxygenase-1 to mitochondria is a novel cytoprotective mechanism against non-steroidal anti-inflammatory drug-induced mitochondrial oxidative stress, apoptosis, and gastric mucosal injury, J Biol Chem, vol.286, pp.39387-402, 2011.

S. Bansal, G. Biswas, and N. G. Avadhani, Mitochondria-targeted heme oxygenase-1 induces oxidative stress and mitochondrial dysfunction in macrophages, kidney fibroblasts and in chronic alcohol hepatotoxicity, Redox Biol, vol.2, pp.273-83, 2014.
DOI : 10.1016/j.redox.2013.07.004

URL : https://doi.org/10.1016/j.redox.2013.07.004

B. Beutler, Innate immunity: an overview, Mol Immunol, vol.40, pp.845-59, 2004.

Y. Delneste, C. Beauvillain, and P. Jeannin, Innate immunity: structure and function of TLRs, Med Sci (Paris), vol.23, pp.67-73, 2007.

K. A. Brown, S. D. Brain, J. D. Pearson, J. D. Edgeworth, S. M. Lewis et al., Neutrophils in development of multiple organ failure in sepsis, Lancet, vol.368, pp.157-69, 2006.

P. Efron and L. L. Moldawer, Sepsis and the dendritic cell, Immunology, vol.20, pp.1-12, 2003.

K. M. Nautiyal, H. Mckellar, A. J. Silverman, and R. Silver, Mast cells are necessary for the hypothermic response to LPS-induced sepsis, Am J Physiol Regul Integr Comp Physiol, vol.296, pp.595-602, 2009.
DOI : 10.1152/ajpregu.90888.2008

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2665855/pdf

A. Hoetzel, T. Dolinay, R. Schmidt, A. M. Choi, and S. W. Ryter, Carbon monoxide in sepsis, Antioxid Redox Signal, vol.9, pp.2013-2039, 2007.

X. Mu, C. Pan, S. Zheng, Y. Alhamdi, B. Sun et al., Protective effects of carbon monoxide-releasing molecule-2 on the barrier function of intestinal epithelial cells, PLoS One, vol.9, p.104032, 2014.

E. H. Warren, X. C. Zhang, S. Li, W. Fan, B. E. Storer et al., Effect of MHC and non-MHC donor/recipient genetic disparity on the outcome of allogeneic HCT, Blood, vol.120, pp.2796-806, 2012.

A. E. Morelli and A. W. Thomson, Tolerogenic dendritic cells and the quest for transplant tolerance, Nat Rev Immunol, vol.7, pp.610-631, 2007.
DOI : 10.1038/nri2132

D. S. Gould and H. Auchincloss, Direct and indirect recognition: the role of MHC antigens in graft rejection, Immunol Today, vol.20, pp.77-82, 1999.

J. M. Ali, E. M. Bolton, J. A. Bradley, and G. J. Pettigrew, Allorecognition pathways in transplant rejection and tolerance, Transplantation, vol.96, pp.681-689, 2013.
DOI : 10.1097/tp.0b013e31829853ce

L. A. Harshyne, S. C. Watkins, A. Gambotto, and S. M. Barratt-boyes, Dendritic cells acquire antigens from live cells for cross-presentation to CTL, J Immunol, vol.166, pp.3717-3740, 2001.
DOI : 10.4049/jimmunol.166.6.3717

URL : http://www.jimmunol.org/content/jimmunol/166/6/3717.full.pdf

G. Benichou and A. W. Thomson, Direct versus indirect allorecognition pathways: on the right track, Am J Transplant, vol.9, pp.655-661, 2009.

U. Schliesser, M. Streitz, and B. Sawitzki, Tregs: application for solid-organ transplantation, Curr Opin Organ Transplant, vol.17, pp.34-41, 2012.

C. Guillonneau, E. Picarda, and I. Anegon, CD8 + regulatory T cells in solid organ transplantation, Curr Opin Organ Transplant, vol.15, pp.751-757, 2010.

E. Picarda, S. Bezie, V. Venturi, K. Echasserieau, E. Merieau et al., MHCderived allopeptide activates TCR-biased CD8
DOI : 10.1172/jci71533

URL : http://www.jci.org/articles/view/71533/files/pdf

, + Tregs and suppresses organ rejection

, Clin Invest, vol.124, pp.2497-512, 2014.

A. Pileggi, R. D. Molano, T. Berney, P. Cattan, C. Vizzardelli et al., Heme oxygenase-1 induction in islet cells results in protection from apoptosis and improved in vivo function after transplantation, Diabetes, vol.50, pp.1983-91, 2001.

C. M. Hu, H. H. Lin, M. T. Chiang, P. F. Chang, and L. Y. Chau, Systemic expression of heme oxygenase-1 ameliorates type 1 diabetes in NOD mice, Diabetes, vol.56, pp.1240-1247, 2007.

J. W. Kupiec-weglinski and R. W. Busuttil, Ischemia and reperfusion injury in liver transplantation, Transplant Proc, vol.37, pp.1653-1659, 2005.

C. Baan, A. Peeters, F. Lemos, A. Uitterlinden, I. Doxiadis et al., Fundamental role for HO-1 in the self-protection of renal allografts, Am J Transplant, vol.4, pp.811-819, 2004.

C. Cai, L. Teng, D. Vu, J. Q. He, Y. Guo et al., The heme oxygenase 1 inducer (CoPP) protects human cardiac stem cells against apoptosis through activation of the extracellular signal-regulated kinase (ERK)/NRF2 signaling pathway and cytokine release, J Biol Chem, vol.287, pp.33720-33752, 2012.

J. F. George, A. Braun, T. M. Brusko, R. Joseph, S. Bolisetty et al., Suppression by CD4 + CD25 + regulatory T cells is dependent on expression of heme oxygenase-1 in antigen-presenting cells, Am J Pathol, vol.173, pp.154-60, 2008.

K. Karimi, N. Kandiah, J. Chau, J. Bienenstock, and P. Forsythe, A Lactobacillus rhamnosus strain induces a heme oxygenase dependent increase in Foxp3 + regulatory T cells, PLoS One, vol.7, p.47556, 2012.

S. Schulz, K. M. Chisholm, H. Zhao, F. Kalish, Y. Yang et al., Heme oxygenase-1 confers protection and alters T-cell populations in a mouse model of neonatal intestinal inflammation, Pediatr Res, vol.77, pp.640-648, 2015.

A. Schumacher and A. C. Zenclussen, Effects of heme oxygenase-1 on innate and adaptive immune responses promoting pregnancy success and allograft tolerance, Front Pharmacol, vol.5, p.288, 2014.

M. Ozen, H. Zhao, D. B. Lewis, R. J. Wong, and D. K. Stevenson, Heme oxygenase and the immune system in normal and pathological pregnancies, Front Pharmacol, vol.6, p.84, 2015.

M. L. Zenclussen, I. Anegon, A. Z. Bertoja, C. Chauveau, K. Vogt et al., Overexpression of heme oxygenase-1 by adenoviral gene transfer improves pregnancy outcome in a murine model of abortion, J Reprod Immunol, vol.69, pp.35-52, 2006.

A. Sollwedel, A. Z. Bertoja, M. L. Zenclussen, K. Gerlof, U. Lisewski et al., Protection from abortion by heme oxygenase-1 up-regulation is associated with increased levels of Bag-1 and neuropilin-1 at the fetal-maternal interface, J Immunol, vol.175, pp.4875-85, 2005.

A. Schumacher, P. O. Wafula, A. Teles, T. El-mousleh, N. Linzke et al., Blockage of heme oxygenase-1 abrogates the protective effect of regulatory T cells on murine pregnancy and promotes the maturation of dendritic cells, PLoS One, vol.7, p.42301, 2012.

M. E. Solano, M. K. Kowal, G. E. O'rourke, A. K. Horst, K. Modest et al., Progesterone and HMOX-1 promote fetal growth by CD8

, + T cell modulation, J Clin Invest, vol.125, pp.1726-1764, 2015.

M. Kwiatek, T. Geca, A. Krzyzanowski, A. Malec, and A. Kwasniewska, Peripheral dendritic cells and CD4 + CD25 + Foxp3 + regulatory T cells in the first trimester of normal pregnancy and in women with recurrent miscarriage, PLoS One, vol.10, p.124747, 2015.

U. Kammerer, A. O. Eggert, M. Kapp, A. D. Mclellan, T. B. Geijtenbeek et al., Unique appearance of proliferating antigen-presenting cells expressing DC-SIGN (CD209) in the decidua of early human pregnancy, Am J Pathol, vol.162, pp.887-96, 2003.

S. M. Blois, A. Soto, C. D. Tometten, M. Klapp, B. F. Margni et al., Lineage, maturity, and phenotype of uterine murine dendritic cells throughout gestation indicate a protective role in maintaining pregnancy, Biol Reprod, vol.70, pp.1018-1041, 2004.

J. R. Prins, B. Zhang, J. E. Schjenken, L. R. Guerin, S. C. Barry et al., Unstable Foxp3 + regulatory T cells and altered dendritic cells are associated with lipopolysaccharide-induced fetal loss in pregnant interleukin 10-deficient mice, Biol Reprod, vol.93, p.95, 2015.

A. C. Zenclussen, D. N. Olivieri, M. L. Dustin, and C. E. Tadokoro, In vivo multiphoton microscopy technique to reveal the physiology of the mouse uterus, Am J Reprod Immunol, vol.69, pp.281-290, 2013.

V. Plaks, T. Birnberg, T. Berkutzki, S. Sela, A. Benyashar et al., Uterine DCs are crucial for decidua formation during embryo implantation in mice, J Clin Invest, vol.118, pp.3954-65, 2008.

M. Tachibana, M. Hashino, T. Nishida, T. Shimizu, and M. Watarai, Protective role of heme oxygenase-1 in Listeria monocytogenes-induced abortion, PLoS One, vol.6, p.25046, 2011.

M. Tachibana, K. Watanabe, Y. Yamasaki, H. Suzuki, and M. Watarai, Expression of heme oxygenase-1 is associated with abortion caused by Brucella abortus infection in pregnant mice, Microb Pathog, vol.45, pp.105-114, 2008.

H. Zhao, R. J. Wong, F. S. Kalish, N. R. Nayak, and D. K. Stevenson, Effect of heme oxygenase-1 deficiency on placental development, Placenta, vol.30, pp.861-869, 2009.

K. Kahlo, S. Malfertheiner, T. Ignatov, F. Jensen, S. D. Costa et al., HO-1 as modulator of the innate immune response in pregnancy, Am J Reprod Immunol, vol.70, pp.24-30, 2013.

N. Linzke, A. Schumacher, K. Woidacki, B. A. Croy, and A. C. Zenclussen, Carbon monoxide promotes proliferation of uterine natural killer cells and remodeling of spiral arteries in pregnant hypertensive heme oxygenase-1 mutant mice, Hypertension, vol.63, pp.580-588, 2014.

M. L. Zenclussen, N. Linzke, A. Schumacher, S. Fest, N. Meyer et al., Heme oxygenase-1 is critically involved in placentation, spiral artery remodeling, and blood pressure regulation during murine pregnancy, Front Pharmacol, vol.5, p.291, 2014.

M. L. Zenclussen, P. A. Casalis, T. El-mousleh, S. Rebelo, S. Langwisch et al., Haem oxygenase-1 dictates intrauterine fetal survival in mice via carbon monoxide, J Pathol, vol.225, pp.293-304, 2011.

T. El-mousleh, P. A. Casalis, I. Wollenberg, M. L. Zenclussen, H. D. Volk et al., Exploring the potential of low doses carbon monoxide as therapy in pregnancy complications, Med Gas Res, vol.2, pp.1-12, 2012.