Immune Netw.  2009 Apr;9(2):46-52. 10.4110/in.2009.9.2.46.

The Role of Nitric Oxide in Mycobacterial Infections

Affiliations
  • 1Department of Microbiology and Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon, Korea. hayoungj@cnu.ac.kr

Abstract

Although tuberculosis poses a significant health threat to the global population, it is a challenge to develop new and effective therapeutic strategies. Nitric oxide (NO) and inducible NO synthase (iNOS) are important in innate immune responses to various intracellular bacterial infections, including mycobacterial infections. It is generally recognized that reactive nitrogen intermediates play an effective role in host defense mechanisms against tuberculosis. In a murine model of tuberculosis, NO plays a crucial role in antimycobacterial activity; however, it is controversial whether NO is critically involved in host defense against Mycobacterium tuberculosis in humans. Here, we review the roles of NO in host defense against murine and human tuberculosis. We also discuss the specific roles of NO in the central nervous system and lung epithelial cells during mycobacterial infection. A greater understanding of these defense mechanisms in human tuberculosis will aid in the development of new strategies for the treatment of disease.

Keyword

nitric oxide; mycobacteria; macrophages; host defense

MeSH Terms

Bacterial Infections
Central Nervous System
Defense Mechanisms
Epithelial Cells
Humans
Immunity, Innate
Lung
Macrophages
Mycobacterium tuberculosis
Nitric Oxide
Nitric Oxide Synthase
Nitrogen
Tuberculosis
Nitric Oxide
Nitric Oxide Synthase
Nitrogen

Figure

  • Figure 1 Yang et al. Synthesis, regulation, and antimycobacterial function of NO in mycobacterial infection. Activated inducible nitric oxide synthase (iNOS) produces Nω-OH-L-arginine from L-arginine, and then Nω-OH-L-arginine is transduced to form NO and L-citruline. Synthesis of NO and reactive nitrogen oxides (RNI) are positively regulated by Th1 cytokines, whereas they are negatively regulated by Th2 cytokines. Produced NO and RNIs, which combined with NO and O2-, can directly kill intracellular MTB in the infected cells (including macrophages, epithelial and glial cells), although the action of NO is dependent on the species and specific cell types.


Reference

1. Adams LB, Dinauer MC, Morgenstern DE, Krahenbuhl JL. Comparison of the roles of reactive oxygen and nitrogen intermediates in the host response to Mycobacterium tuberculosis using transgenic mice. Tuber Lung Dis. 1997. 78:237–246.
Article
2. Chan J, Xing Y, Magliozzo RS, Bloom BR. Killing of virulent Mycobacterium tuberculosis by reactive nitrogen intermediates produced by activated murine macrophages. J Exp Med. 1992. 175:1111–1122.
Article
3. Rojas M, Barrera LF, Puzo G, Garcia LF. Differential induction of apoptosis by virulent Mycobacterium tuberculosis in resistant and susceptible murine macrophages: role of nitric oxide and mycobacterial products. J Immunol. 1997. 159:1352–1361.
4. Flynn JL, Scanga CA, Tanaka KE, Chan J. Effects of aminoguanidine on latent murine tuberculosis. J Immunol. 1998. 160:1796–1803.
5. Nathan C, Shiloh MU. Reactive oxygen and nitrogen intermediates in the relationship between mammalian hosts and microbial pathogens. Proc Natl Acad Sci U S A. 2000. 97:8841–8848.
Article
6. Bermudez LE. Differential mechanisms of intracellular killing of Mycobacterium avium and Listeria monocytogenes by activated human and murine macrophages. The role of nitric oxide. Clin Exp Immunol. 1993. 91:277–281.
Article
7. Chan J, Tanaka K, Carroll D, Flynn J, Bloom BR. Effects of nitric oxide synthase inhibitors on murine infection with Mycobacterium tuberculosis. Infect Immun. 1995. 63:736–740.
Article
8. Kuo HP, Wang CH, Huang KS, Lin HC, Yu CT, Liu CY, Lu LC. Nitric oxide modulates interleukin-1beta and tumor necrosis factor-alpha synthesis by alveolar macrophages in pulmonary tuberculosis. Am J Respir Crit Care Med. 2000. 161:192–199.
Article
9. Lee SC, Dickson DW, Liu W, Brosnan CF. Induction of nitric oxide synthase activity in human astrocytes by interleukin-1beta and interferon-gamma. J Neuroimmunol. 1993. 46:19–24.
Article
10. Wang CH, Lin HC, Liu CY, Huang KH, Huang TT, Yu CT, Kuo HP. Upregulation of inducible nitric oxide synthase and cytokine secretion in peripheral blood monocytes from pulmonary tuberculosis patients. Int J Tuberc Lung Dis. 2001. 5:283–291.
11. Adams LB, Franzblau SG, Vavrin Z, Hibbs JB Jr, Krahenbuhl JL. L-arginine-dependent macrophage effector functions inhibit metabolic activity of Mycobacterium leprae. J Immunol. 1991. 147:1642–1646.
12. Alam MS, Akaike T, Okamoto S, Kubota T, Yoshitake J, Sawa T, Miyamoto Y, Tamura F, Maeda H. Role of nitric oxide in host defense in murine salmonellosis as a function of its antibacterial and antiapoptotic activities. Infect Immun. 2002. 70:3130–3142.
Article
13. MacMicking J, Xie QW, Nathan C. Nitric oxide and macrophage function. Annu Rev Immunol. 1997. 15:323–350.
Article
14. Mayer B, Hemmens B. Biosynthesis and action of nitric oxide in mammalian cells. Trends Biochem Sci. 1997. 22:477–481.
Article
15. Choi HS, Rai PR, Chu HW, Cool C, Chan ED. Analysis of nitric oxide synthase and nitrotyrosine expression in human pulmonary tuberculosis. Am J Respir Crit Care Med. 2002. 166:178–186.
Article
16. Nicholson S, Bonecini-Almeida Mda G, Lapa e Silva JR, Nathan C, Xie QW, Mumford R, Weidner JR, Calaycay J, Geng J, Boechat N, Linhares C, Rom W, Ho JL. Inducible nitric oxide synthase in pulmonary alveolar macrophages from patients with tuberculosis. J Exp Med. 1996. 183:2293–2302.
Article
17. Denis M. In vivo modulation of atypical mycobacterial infection: adjuvant therapy increases resistance to Mycobacterium avium by enhancing macrophage effector functions. Cell Immunol. 1991. 134:42–53.
Article
18. Rodriguez PC, Zea AH, DeSalvo J, Culotta KS, Zabaleta J, Quiceno DG, Ochoa JB, Ochoa AC. L-arginine consumption by macrophages modulates the expression of CD3zeta chain in T lymphocytes. J Immunol. 2003. 171:1232–1239.
Article
19. Robbins RA, Barnes PJ, Springall DR, Warren JB, Kwon OJ, Buttery LD, Wilson AJ, Geller DA, Polak JM. Expression of inducible nitric oxide in human lung epithelial cells. Biochem Biophys Res Commun. 1994. 203:209–218.
Article
20. Paton NI, Chua YK, Earnest A, Chee CB. Randomized controlled trial of nutritional supplementation in patients with newly diagnosed tuberculosis and wasting. Am J Clin Nutr. 2004. 80:460–465.
Article
21. Wu G, Morris SM Jr. Arginine metabolism: nitric oxide and beyond. Biochem J. 1998. 336:1–17.
Article
22. Moncada S, Palmer RM, Higgs EA. Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol Rev. 1991. 43:109–142.
23. Nathan C, Xie QW. Regulation of biosynthesis of nitric oxide. J Biol Chem. 1994. 269:13725–13728.
Article
24. Zaki MH, Akuta T, Akaike T. Nitric oxide-induced nitrative stress involved in microbial pathogenesis. J Pharmacol Sci. 2005. 98:117–129.
Article
25. Ralph AP, Kelly PM, Anstey NM. L-arginine and vitamin D: novel adjunctive immunotherapies in tuberculosis. Trends Microbiol. 2008. 16:336–344.
Article
26. Gow AJ, Thom SR, Ischiropoulos H. Nitric oxide and peroxynitrite-mediated pulmonary cell death. Am J Physiol. 1998. 274:L112–L118.
27. Canthaboo C, Xing D, Wei XQ, Corbel MJ. Investigation of role of nitric oxide in protection from Bordetella pertussis respiratory challenge. Infect Immun. 2002. 70:679–684.
Article
28. Umezawa K, Akaike T, Fujii S, Suga M, Setoguchi K, Ozawa A, Maeda H. Induction of nitric oxide synthesis and xanthine oxidase and their roles in the antimicrobial mechanism against Salmonella typhimurium infection in mice. Infect Immun. 1997. 65:2932–2940.
Article
29. Zaragoza C, Ocampo C, Saura M, Leppo M, Wei XQ, Quick R, Moncada S, Liew FY, Lowenstein CJ. The role of inducible nitric oxide synthase in the host response to coxsackievirus myocarditis. Proc Natl Acad Sci U S A. 1998. 95:2469–2474.
Article
30. Zaragoza C, Ocampo CJ, Saura M, Bao C, Leppo M, Lafond-Walker A, Thiemann DR, Hruban R, Lowenstein CJ. Inducible nitric oxide synthase protection against coxsackievirus pancreatitis. J Immunol. 1999. 163:5497–5504.
31. Zaragoza C, Ocampo CJ, Saura M, McMillan A, Lowenstein CJ. Nitric oxide inhibition of coxsackievirus replication in vitro. J Clin Invest. 1997. 100:1760–1767.
Article
32. Mannick JB, Asano K, Izumi K, Kieff E, Stamler JS. Nitric oxide produced by human B lymphocytes inhibits apoptosis and Epstein-Barr virus reactivation. Cell. 1994. 79:1137–1146.
Article
33. Croen KD. Evidence for antiviral effect of nitric oxide. Inhibition of herpes simplex virus type 1 replication. J Clin Invest. 1993. 91:2446–2452.
Article
34. Gamba G, Cavalieri H, Courreges MC, Massouh EJ, Benencia F. Early inhibition of nitric oxide production increases HSV-1 intranasal infection. J Med Virol. 2004. 73:313–322.
Article
35. MacLean A, Wei XQ, Huang FP, Al-Alem UA, Chan WL, Liew FY. Mice lacking inducible nitric-oxide synthase are more susceptible to herpes simplex virus infection despite enhanced Th1 cell responses. J Gen Virol. 1998. 79:825–830.
Article
36. Akaike T, Okamoto S, Sawa T, Yoshitake J, Tamura F, Ichimori K, Miyazaki K, Sasamoto K, Maeda H. 8-nitroguanosine formation in viral pneumonia and its implication for pathogenesis. Proc Natl Acad Sci U S A. 2003. 100:685–690.
Article
37. Kreil TR, Eibl MM. Nitric oxide and viral infection: NO antiviral activity against a flavivirus in vitro, and evidence for contribution to pathogenesis in experimental infection in vivo. Virology. 1996. 219:304–306.
Article
38. Yoshitake J, Akaike T, Akuta T, Tamura F, Ogura T, Esumi H, Maeda H. Nitric oxide as an endogenous mutagen for Sendai virus without antiviral activity. J Virol. 2004. 78:8709–8719.
Article
39. Adler H, Beland JL, Del-Pan NC, Kobzik L, Brewer JP, Martin TR, Rimm IJ. Suppression of herpes simplex virus type 1 (HSV-1)-induced pneumonia in mice by inhibition of inducible nitric oxide synthase (iNOS, NOS2). J Exp Med. 1997. 185:1533–1540.
Article
40. Fujii S, Akaike T, Maeda H. Role of nitric oxide in pathogenesis of herpes simplex virus encephalitis in rats. Virology. 1999. 256:203–212.
Article
41. Bolovan-Fritts CA, Spector SA. Endothelial damage from cytomegalovirus-specific host immune response can be prevented by targeted disruption of fractalkine-CX3CR1 interaction. Blood. 2008. 111:175–182.
Article
42. Zhang M, Xin H, Atherton SS. Murine cytomegalovirus (MCMV) spreads to and replicates in the retina after endotoxin-induced disruption of the blood-retinal barrier of immunosuppressed BALB/c mice. J Neurovirol. 2005. 11:365–375.
Article
43. MacMicking JD, North RJ, LaCourse R, Mudgett JS, Shah SK, Nathan CF. Identification of nitric oxide synthase as a protective locus against tuberculosis. Proc Natl Acad Sci U S A. 1997. 94:5243–5248.
Article
44. Arias M, Rojas M, Zabaleta J, Rodríguez JI, París SC, Barrera LF, García LF. Inhibition of virulent Mycobacterium tuberculosis by Bcg(r) and Bcg(s) macrophages correlates with nitric oxide production. J Infect Dis. 1997. 176:1552–1558.
Article
45. Flesch IE, Hess JH, Kaufmann SH. NADPH diaphorase staining suggests a transient and localized contribution of nitric oxide to host defence against an intracellular pathogen in situ. Int Immunol. 1994. 6:1751–1757.
Article
46. Doi T, Ando M, Akaike T, Suga M, Sato K, Maeda H. Resistance to nitric oxide in Mycobacterium avium complex and its implication in pathogenesis. Infect Immun. 1993. 61:1980–1989.
Article
47. Scanga CA, Mohan VP, Yu K, Joseph H, Tanaka K, Chan J, Flynn JL. Depletion of CD4(+) T cells causes reactivation of murine persistent tuberculosis despite continued expression of interferon gamma and nitric oxide synthase 2. J Exp Med. 2000. 192:347–358.
Article
48. Aston C, Rom WN, Talbot AT, Reibman J. Early inhibition of mycobacterial growth by human alveolar macrophages is not due to nitric oxide. Am J Respir Crit Care Med. 1998. 157:1943–1950.
Article
49. Jagannath C, Actor JK, Hunter RL Jr. Induction of nitric oxide in human monocytes and monocyte cell lines by Mycobacterium tuberculosis. Nitric Oxide. 1998. 2:174–186.
Article
50. Kwon OJ. The role of nitric oxide in the immune response of tuberculosis. J Korean Med Sci. 1997. 12:481–487.
Article
51. Rich EA, Torres M, Sada E, Finegan CK, Hamilton BD, Toossi Z. Mycobacterium tuberculosis (MTB)-stimulated production of nitric oxide by human alveolar macrophages and relationship of nitric oxide production to growth inhibition of MTB. Tuber Lung Dis. 1997. 78:247–255.
Article
52. Rockett KA, Brookes R, Udalova I, Vidal V, Hill AV, Kwiatkowski D. 1,25-Dihydroxyvitamin D3 induces nitric oxide synthase and suppresses growth of Mycobacterium tuberculosis in a human macrophage-like cell line. Infect Immun. 1998. 66:5314–5321.
Article
53. Nozaki Y, Hasegawa Y, Ichiyama S, Nakashima I, Shimokata K. Mechanism of nitric oxide-dependent killing of Mycobacterium bovis BCG in human alveolar macrophages. Infect Immun. 1997. 65:3644–3647.
Article
54. Wang CH, Liu CY, Lin HC, Yu CT, Chung KF, Kuo HP. Increased exhaled nitric oxide in active pulmonary tuberculosis due to inducible NO synthase upregulation in alveolar macrophages. Eur Respir J. 1998. 11:809–815.
Article
55. Facchetti F, Vermi W, Fiorentini S, Chilosi M, Caruso A, Duse M, Notarangelo LD, Badolato R. Expression of inducible nitric oxide synthase in human granulomas and histiocytic reactions. Am J Pathol. 1999. 154:145–152.
Article
56. Kropf P, Baud D, Marshall SE, Munder M, Mosley A, Fuentes JM, Bangham CR, Taylor GP, Herath S, Choi BS, Soler G, Teoh T, Modolell M, Muller I. Arginase activity mediates reversible T cell hyporesponsiveness in human pregnancy. Eur J Immunol. 2007. 37:935–945.
Article
57. Gazzinelli RT, Eltoum I, Wynn TA, Sher A. Acute cerebral toxoplasmosis is induced by in vivo neutralization of TNF-alpha and correlates with the down-regulated expression of inducible nitric oxide synthase and other markers of macrophage activation. J Immunol. 1993. 151:3672–3681.
58. Tucker PC, Griffin DE, Choi S, Bui N, Wesselingh S. Inhibition of nitric oxide synthesis increases mortality in Sindbis virus encephalitis. J Virol. 1996. 70:3972–3977.
Article
59. Rock RB, Gekker G, Hu S, Sheng WS, Cheeran M, Lokensgard JR, Peterson PK. Role of microglia in central nervous system infections. Clin Microbiol Rev. 2004. 17:942–964.
Article
60. Rock RB, Hu S, Deshpande A, Munir S, May BJ, Baker CA, Peterson PK, Kapur V. Transcriptional response of human microglial cells to interferon-γ. Genes Immun. 2005. 6:712–719.
Article
61. Peterson PK, Hu S, Anderson WR, Chao CC. Nitric oxide production and neurotoxicity mediated by activated microglia from human versus mouse brain. J Infect Dis. 1994. 170:457–460.
Article
62. Mazzolla R, Puliti M, Barluzzi R, Neglia R, Bistoni F, Barbolini G, Blasi E. Differential microbial clearance and immunoresponse of Balb/c (Nramp1 susceptible) and DBA2 (Nramp1 resistant) mice intracerebrally infected with Mycobacterium bovis BCG (BCG). FEMS Immunol Med Microbiol. 2002. 32:149–158.
Article
63. van Well GT, Wieland CW, Florquin S, Roord JJ, van der Poll T, van Furth AM. A new murine model to study the pathogenesis of tuberculous meningitis. J Infect Dis. 2007. 195:694–697.
Article
64. Olin MR, Armien AG, Cheeran MC, Rock RB, Molitor TW, Peterson PK. Role of nitric oxide in defense of the central nervous system against Mycobacterium tuberculosis. J Infect Dis. 2008. 198:886–889.
Article
65. Kwon OJ, Kim JH, Kim HC, Suh GY, Park JW, Chung MP, Kim H, Rhee CH. Nitric oxide expression in airway epithelial cells in response to tubercle bacilli stimulation. Respirology. 1998. 3:119–124.
Article
66. Roy S, Sharma S, Sharma M, Aggarwal R, Bose M. Induction of nitric oxide release from the human alveolar epithelial cell line A549: an in vitro correlate of innate immune response to Mycobacterium tuberculosis. Immunology. 2004. 112:471–480.
Article
67. Sharma M, Sharma S, Roy S, Varma S, Bose M. Pulmonary epithelial cells are a source of interferon-gamma in response to Mycobacterium tuberculosis infection. Immunol Cell Biol. 2007. 85:229–237.
Article
Full Text Links
  • IN
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr