Invited Review
The role of TNF in parasitic diseases: Still more questions than answers

https://doi.org/10.1016/j.ijpara.2010.03.011Get rights and content

Abstract

The inhibition of TNF with therapeutic monoclonal antibodies or antibody/receptor fusion proteins in rheumatoid arthritis still constitutes the benchmark for a successful intervention in an ongoing auto-immune-inflammatory disease and underlines the importance of this cytokine. TNF plays a central role in the defence against intracellular infections and is responsible for the promotion of different aspects of the innate immune response such as inflammatory cell recruitment and cell differentiation. While this cytokine generally displays pro-inflammatory activities supporting the early stages of the inflammatory response, it has been demonstrated to be especially important during infection with intracellular pathogens and, consequently, leishmaniasis of TNF−/− mice ends fatally. However, the specific activities of TNF that confer protection are not yet fully understood. This review will summarize the current understanding of TNF function and signalling, and will discuss recent work in the models of malaria, toxoplasmosis, trypanosomiasis and leishmaniasis with particular emphasis on work with gene-deficient mouse models.

Section snippets

TNF and its two receptors

TNF was first named in 1975 as an endotoxin-induced factor that effected the necrosis of established tumors (Carswell et al., 1975). The biology of TNF was further characterised over the next 10 years in two parallel lines of research that studied two fundamentally different metabolic activities of this cytokine: its ability to induce cell death in certain transformed cell lines (Old, 1985, Espevik and Nissen-Meyer, 1986) and its suppression of the enzyme lipoprotein lipase A (Kawakami et al.,

TNF and its receptors in malaria

Symptomatic malaria infection arises when the causative parasite, Plasmodium spp., invades and replicates within circulating erythrocytes, frequently causing relatively mild symptoms such as nausea and fever, but occasionally more severe problems such as anaemia, respiratory distress and cerebral malaria (CM). In CM Plasmodium falciparum-infected erythrocytes cyto-adhere to the brain microvasculature, resulting in a massive functional disturbance of the brain that is often life threatening.

Concluding remarks

The role of TNF has been investigated in many experimental models of infection. The results which have been reported are not always consistent and sometimes surprisingly contradictory. This review highlights that the genomic make-up of knockout models used to study TNF or TNFR could be a contributing factor to the different outcomes in experimental models of intracellular infection, keeping in mind that back-crossing five generations leaves approximately 3% of non-linked genes from the founder

Acknowledgements

The authors wish to acknowledge the financial support of the NHMRC, the DFG (Schl 392/6-1 to DS) and the Medical Genetics Research Advancement Program (JCU to HK).

References (153)

  • M. Grell et al.

    The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor

    Cell

    (1995)
  • M. Hernandez-Valladares et al.

    Pathology of Tnf-deficient mice infected with Plasmodium chabaudi adami 408XZ

    Exp. Parasitol.

    (2006)
  • M. Higuchi et al.

    Membrane tumor necrosis factor-α (TNF-α) expressed on HTLV-I-infected T cells mediates a costimulatory signal for B cell activation–characterization of membrane TNF-α

    Clin. Immunol. Immunopath.

    (1997)
  • M. Kriegler et al.

    A novel form of TNF/cachectin is a cell surface cytotoxic transmembrane protein: ramifications of the complex physiology of TNF

    Cell

    (1988)
  • D. Kwiatkowski et al.

    TNF concentration in fatal cerebral, non-fatal cerebral, and uncomplicated Plasmodium falciparum malaria

    Lancet

    (1990)
  • S. Lassoued et al.

    Toxoplasmic chorioretinitis and antitumor necrosis factor treatment in rheumatoid arthritis

    Semin. Arthritis Rheum.

    (2007)
  • G.H. Long et al.

    Blockade of TNF receptor 1 reduces disease severity but increases parasite transmission during Plasmodium chabaudi chabaudi infection

    Int. J. Parasitol.

    (2008)
  • R. Lucas et al.

    Respective role of TNF receptors in the development of experimental cerebral malaria

    J. Neuroimmunol.

    (1997)
  • C.L. Mackintosh et al.

    Clinical features and pathogenesis of severe malaria

    Trends Parasitol.

    (2004)
  • M.F. McDermott et al.

    Germline mutations in the extracellular domains of the 55 kDa TNF receptor, TNFR1, define a family of dominantly inherited autoinflammatory syndromes

    Cell

    (1999)
  • W. McGuire et al.

    Levels of tumour necrosis factor and soluble TNF receptors during malaria fever episodes in the community

    Trans. R. Soc. Trop. Med. Hyg.

    (1998)
  • J.G. Montoya et al.

    Toxoplasmosis

    Lancet

    (2004)
  • C. Mueller et al.

    Noncleavable transmembrane mouse tumor necrosis factor-α (TNFα) mediates effects distinct from those of wild-type TNFα in vitro and in vivo

    J. Biol. Chem.

    (1999)
  • M.A. Munoz-Fernandez et al.

    Activation of human macrophages for the killing of intracellular Trypanosoma cruzi by TNF-α and IFN-γ through a nitric oxide-dependent mechanism

    Immunol. Lett.

    (1992)
  • J.R. Muppidi et al.

    Life and death decisions: secondary complexes and lipid rafts in TNF receptor family signal transduction

    Immunity

    (2004)
  • C. Allenbach et al.

    An essential role for transmembrane TNF in the resolution of the inflammatory lesion induced by Leishmania major infection

    Eur. J. Immunol.

    (2008)
  • G. Aversa et al.

    The 26-kD transmembrane form of tumor necrosis factor α on activated CD4+ T cell clones provides a costimulatory signal for human B cell activation

    J. Exp. Med.

    (1993)
  • F. Bazzoni et al.

    The tumor necrosis factor ligand and receptor families

    N. Engl. J. Med.

    (1996)
  • J.H. Bell et al.

    Role of ADAM17 in the ectodomain shedding of TNF-α and its receptors by neutrophils and macrophages

    J. Leukoc. Biol.

    (2007)
  • S. Bennouna et al.

    Cross-talk in the innate immune system: neutrophils instruct recruitment and activation of dendritic cells during microbial infection

    J. Immunol.

    (2003)
  • B. Beutler et al.

    Identity of tumour necrosis factor and the macrophage-secreted factor cachectin

    Nature

    (1985)
  • B. Beutler et al.

    Purification of cachectin, a lipoprotein lipase-suppressing hormone secreted by endotoxin-induced RAW 264.7 cells

    J. Exp. Med.

    (1985)
  • C.M. Black et al.

    Effect of recombinant tumour necrosis factor on acute infection in mice with Toxoplasma gondii or Trypanosoma cruzi

    Immunology

    (1989)
  • R.A. Black et al.

    A metalloproteinase disintegrin that releases tumour-necrosis factor-α from cells

    Nature

    (1997)
  • M. Brockhaus et al.

    Identification of two types of tumor necrosis factor receptors on human cell lines by monoclonal antibodies

    Proc. Natl. Acad. Sci. USA

    (1990)
  • C.R. Brown et al.

    Definitive identification of a gene that confers resistance against Toxoplasma cyst burden and encephalitis

    Immunology

    (1995)
  • H. Brown et al.

    Cytokine expression in the brain in human cerebral malaria

    J. Infect. Dis.

    (1999)
  • M.M. Camargo et al.

    Glycosylphosphatidylinositol-anchored mucin-like glycoproteins isolated from Trypanosoma cruzi trypomastigotes initiate the synthesis of proinflammatory cytokines by macrophages

    J. Immunol.

    (1997)
  • E.A. Carswell et al.

    An endotoxin-induced serum factor that causes necrosis of tumors

    Proc. Natl. Acad. Sci. USA

    (1975)
  • E. Castanos-Velez et al.

    Trypanosoma cruzi infection in tumor necrosis factor receptor p55-deficient mice

    Infect. Immun.

    (1998)
  • I.A. Clark et al.

    Inhibition of murine malaria (Plasmodium chabaudi) in vivo by recombinant interferon-gamma or tumor necrosis factor, and its enhancement by butylated hydroxyanisole

    J. Immunol.

    (1987)
  • K. Clark et al.

    Lymphotoxin α and tumour necrosis factor are not required for control of parasite growth, but differentially regulate cytokine production during Plasmodium chabaudi chabaudi AS infection

    Parasite Immunol.

    (2007)
  • P.D. Crowe et al.

    A lymphotoxin-β-specific receptor

    Science

    (1994)
  • S. Daulouede et al.

    Human macrophage tumor necrosis factor (TNF)-α production induced by Trypanosoma brucei gambiense and the role of TNF-α in parasite control

    J. Infect. Dis.

    (2001)
  • S. de Kossodo et al.

    Tumor necrosis factor α (TNF-α) and TNF-β and their receptors in experimental cutaneous leishmaniasis

    Infect. Immun.

    (1994)
  • F. De Leonardis et al.

    Visceral leishmaniasis and anti-TNF-α therapy: case report and review of the literature

    Clin. Exp. Rheumatol.

    (2009)
  • J.B. de Souza et al.

    Cerebral malaria: the contribution of studies in animal models to our understanding of immunopathogenesis

    Microbes Infect.

    (2002)
  • F. Debierre-Grockiego et al.

    Activation of TLR2 and TLR4 by glycosylphosphatidylinositols derived from Toxoplasma gondii

    J. Immunol.

    (2007)
  • F. Debierre-Grockiego et al.

    Fatty acids isolated from Toxoplasma gondii reduce glycosylphosphatidylinositol-induced tumor necrosis factor α production through inhibition of the NF-κB signaling pathway

    Infect. Immun.

    (2007)
  • M. Deckert-Schluter et al.

    Toxoplasma encephalitis in congenic B10 and BALB mice: impact of genetic factors on the immune response

    Infect. Immun.

    (1994)
  • Cited by (0)

    View full text