Heme oxygenase-1 affords protection against noncerebral forms of severe malaria

Infection by Plasmodium, the causative agent of malaria, is associated with hemolysis and therefore with release of hemoglobin from RBC. Under inflammatory conditions, cell-free hemoglobin can be oxidized, releasing its heme prosthetic groups and producing deleterious free heme. Here we demonstrate that survival of a Plasmodium-infected host relies strictly on its ability to prevent the cytotoxic effects of free heme via the expression of the heme-catabolyzing enzyme heme oxygenase-1 (HO-1; encoded by the Hmox1 gene). When infected with Plasmodium chabaudi chabaudi (Pcc), wild-type (Hmox1+/+) BALB/c mice resolved infection and restored homeostasis thereafter (0% lethality). In contrast, HO-1 deficient (Hmox1−/−) BALB/c mice developed a lethal form of hepatic failure (100% lethality), similar to the one occurring in Pcc-infected DBA/2 mice (75% lethality). Expression of HO-1 suppresses the pro-oxidant effects of free heme, preventing it from sensitizing hepatocytes to undergo TNF-mediated programmed cell death by apoptosis. This cytoprotective effect, which inhibits the development of hepatic failure in Pcc-infected mice without interfering with pathogen burden, is mimicked by pharmacological antioxidants such as N-acetylcysteine (NAC). When administered therapeutically, i.e., after Pcc infection, NAC suppressed the development of hepatic failure in Pcc-infected DBA/2 mice (0% lethality), without interfering with pathogen burden. In conclusion, we describe a mechanism of host defense against Plasmodium infection, based on tissue cytoprotection against free heme and limiting disease severity irrespectively of parasite burden.

[1]  F. Bach,et al.  Heme oxygenase-1: from biology to therapeutic potential. , 2009, Trends in molecular medicine.

[2]  David S Schneider,et al.  Two ways to survive infection: what resistance and tolerance can teach us about treating infectious diseases , 2008, Nature Reviews Immunology.

[3]  Melissa D. Laird,et al.  Hemin-induced necroptosis involves glutathione depletion in mouse astrocytes. , 2008, Free radical biology & medicine.

[4]  M. Soares,et al.  A central role for free heme in the pathogenesis of severe malaria: the missing link? , 2008, Journal of Molecular Medicine.

[5]  R. Medzhitov Origin and physiological roles of inflammation , 2008, Nature.

[6]  Elsa Seixas,et al.  An experimental model for fatal malaria due to TNF-α-dependent hepatic damage , 2008, Parasitology.

[7]  L. Gonçalves,et al.  Improved isolation of murine hepatocytes for in vitro malaria liver stage studies , 2007, Malaria Journal.

[8]  D. Sim,et al.  Disentangling Genetic Variation for Resistance and Tolerance to Infectious Diseases in Animals , 2007, Science.

[9]  M. Mota,et al.  Heme oxygenase-1 and carbon monoxide suppress the pathogenesis of experimental cerebral malaria , 2007, Nature Medicine.

[10]  P. Fontoura,et al.  Heme oxygenase-1 and carbon monoxide suppress autoimmune neuroinflammation. , 2007, The Journal of clinical investigation.

[11]  V. Choubey,et al.  Apoptosis in liver during malaria: role of oxidative stress and implication of mitochondrial pathway , 2006, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[12]  S. Hay,et al.  The global distribution of clinical episodes of Plasmodium falciparum malaria , 2005, Nature.

[13]  T. Strom,et al.  Heme oxygenase‐1 modulates the allo‐immune response by promoting activation‐induced cell death of T cells , 2005, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[14]  I. Clark,et al.  Pathogenesis of Malaria and Clinically Similar Conditions , 2004, Clinical Microbiology Reviews.

[15]  B. Aggarwal Signalling pathways of the TNF superfamily: a double-edged sword , 2003, Nature Reviews Immunology.

[16]  G. Brittenham,et al.  N-acetylcysteine in severe falciparum malaria in Thailand. , 2003, The Southeast Asian journal of tropical medicine and public health.

[17]  G. Watt,et al.  A pilot study of N-acetylcysteine as adjunctive therapy for severe malaria. , 2002, QJM : monthly journal of the Association of Physicians.

[18]  Ogobara K. Doumbo,et al.  The pathogenic basis of malaria , 2002, Nature.

[19]  L. Kobzik,et al.  Hypoxia induces severe right ventricular dilatation and infarction in heme oxygenase-1 null mice. , 1999, The Journal of clinical investigation.

[20]  J. Langhorne,et al.  gammadelta T cells contribute to control of chronic parasitemia in Plasmodium chabaudi infections in mice. , 1999, Journal of immunology.

[21]  D. Warrell,et al.  Neurological manifestations of falciparum malaria , 1998, Annals of neurology.

[22]  R. Snow,et al.  Indicators of life-threatening malaria in African children. , 1995, The New England journal of medicine.

[23]  J. Eaton,et al.  Endothelial-cell heme uptake from heme proteins: induction of sensitization and desensitization to oxidant damage. , 1993, Proceedings of the National Academy of Sciences of the United States of America.

[24]  Mike Rothe,et al.  Tumor necrosis factor's cytotoxic activity is signaled by the p55 TNF receptor , 1993, Cell.

[25]  H. Marver,et al.  The enzymatic conversion of heme to bilirubin by microsomal heme oxygenase. , 1968, Proceedings of the National Academy of Sciences of the United States of America.

[26]  A. Bhalla,et al.  Malarial hepatopathy. , 2006, Journal of postgraduate medicine.