Plasmodium yoelii 17XL infection up-regulates RANTES, CCR1, CCR3 and CCR5 expression, and induces ultrastructural changes in the cerebellum

Malar J. 2005 Dec 16:4:63. doi: 10.1186/1475-2875-4-63.

Abstract

Background: Malaria afflicts 300-500 million people causing over 1 million deaths globally per year. The immunopathogenesis of malaria is mediated partly by co mplex cellular and immunomodulator interactions involving co-regulators such as cytokines and adhesion molecules. However, the role of chemokines and their receptors in malaria immunopathology remains unclear. RANTES (Regulated on Activation Normal T-Cell Expressed and Secreted) is a chemokine involved in the generation of inflammatory infiltrates. Recent studies indicate that the degradation of cell-cell junctions, blood-brain barrier dysfunction, recruitment of leukocytes and Plasmodium-infected erythrocytes into and occlusion of microvessels relevant to malaria pathogenesis are associated with RANTES expression. Additionally, activated lymphocytes, platelets and endothelial cells release large quantities of RANTES, thus suggesting a unique role for RANTES in the generation and maintenance of the malaria-induced inflammatory response. The hypothesis of this study is that RANTES and its corresponding receptors (CCR1, CCR3 and CCR5) modulate malaria immunopathogenesis. A murine malaria model was utilized to evaluate the role of this chemokine and its receptors in malaria.

Methods: The alterations in immunomodulator gene expression in brains of Plasmodium yoelii 17XL-infected mice was analysed using cDNA microarray screening, followed by a temporal comparison of mRNA and protein expression of RANTES and its corresponding receptors by qRT-PCR and Western blot analysis, respectively. Plasma RANTES levels was determined by ELISA and ultrastructural studies of brain sections from infected and uninfected mice was conducted.

Results: RANTES (p < 0.002), CCR1 (p < 0.036), CCR3 (p < 0.033), and CCR5 (p < 0.026) mRNA were significantly upregulated at peak parasitaemia and remained high thereafter in the experimental mouse model. RANTES protein in the brain of infected mice was upregulated (p < 0.034) compared with controls. RANTES plasma levels were significantly upregulated; two to three fold in infected mice compared with controls (p < 0.026). Some distal microvascular endothelium in infected cerebellum appeared degraded, but remained intact in controls.

Conclusion: The upregulation of RANTES, CCR1, CCR3, and CCR5 mRNA, and RANTES protein mediate inflammation and cellular degradation in the cerebellum during P. yoelii 17XL malaria.

Publication types

  • Comparative Study
  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Blotting, Western / methods
  • Cerebellum / pathology
  • Cerebellum / ultrastructure*
  • Chemokine CCL5 / biosynthesis*
  • Chemokine CCL5 / physiology
  • DNA Primers / chemistry
  • Female
  • Gene Expression Profiling / methods
  • Humans
  • Immunologic Factors / physiology
  • Malaria / immunology*
  • Malaria / pathology
  • Mice
  • Oligonucleotide Array Sequence Analysis / methods
  • Plasmodium yoelii* / growth & development
  • Plasmodium yoelii* / pathogenicity
  • Receptors, CCR1
  • Receptors, CCR3
  • Receptors, CCR5 / biosynthesis
  • Receptors, CCR5 / physiology
  • Receptors, Chemokine / biosynthesis*
  • Receptors, Chemokine / physiology
  • Reverse Transcriptase Polymerase Chain Reaction / methods
  • Time Factors
  • Up-Regulation / genetics
  • Up-Regulation / physiology

Substances

  • CCR1 protein, human
  • CCR3 protein, human
  • Ccr1 protein, mouse
  • Ccr3 protein, mouse
  • Chemokine CCL5
  • DNA Primers
  • Immunologic Factors
  • Receptors, CCR1
  • Receptors, CCR3
  • Receptors, CCR5
  • Receptors, Chemokine