Genistein-Supplemented Diet Decreases Malaria Liver Infection in Mice and Constitutes a Potential Prophylactic Strategy

In tropical regions millions of people still live at risk of malaria infection. Indeed the emergence of resistance to chloroquine and other drugs in use in these areas reinforces the need to implement alternative prophylactic strategies. Genistein is a naturally occurring compound that is widely used as a food supplment and is thought to be effective in countering several pathologies. Results presented here show that genistein inhibits liver infection by the Plasmodium parasite, the causative agent of malaria. In vitro, genistein decreased the infection rates of both mouse and human hepatoma cells by inhibiting the early stages of the parasite's intracellular development. Oral or intraperitoneal administration of genistein decreased the liver parasite load of P. berghei-infected mice. Moreover, mice fed on a genistein-supplemented diet showed a significant reduction in Plasmodium liver infection as well as a reduced blood parasitemia and partial protection from severe disease. Since genistein is a safe, low-cost, natural compound that can be used permanently in a diet, we propose its use as a prophylactic agent against malaria for endemic populations and long-time travelers.

[1]  Q. Bassat,et al.  Safety of the RTS,S/AS02D candidate malaria vaccine in infants living in a highly endemic area of Mozambique: a double blind randomised controlled phase I/IIb trial , 2007, The Lancet.

[2]  C. Klein,et al.  Genistein genotoxicity: critical considerations of in vitro exposure dose. , 2007, Toxicology and applied pharmacology.

[3]  M. Mota,et al.  Dissecting in vitro host cell infection by Plasmodium sporozoites using flow cytometry , 2007, Cellular microbiology.

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

[5]  Ana Rodriguez,et al.  The silent path to thousands of merozoites: the Plasmodium liver stage , 2006, Nature Reviews Microbiology.

[6]  M. Mota,et al.  Antimalarial drugs – host targets (re)visited , 2006, Biotechnology journal.

[7]  A. C. Alves,et al.  Malaria Parasites Can Develop Stable Resistance to Artemisinin but Lack Mutations in Candidate Genes atp6 (Encoding the Sarcoplasmic and Endoplasmic Reticulum Ca2+ ATPase), tctp, mdr1, and cg10 , 2006, Antimicrobial Agents and Chemotherapy.

[8]  K. Marsh,et al.  Immune effector mechanisms in malaria , 2006, Parasite immunology.

[9]  Oliver Billker,et al.  Protein kinases as targets for antimalarial intervention: Kinomics, structure-based design, transmission-blockade, and targeting host cell enzymes. , 2005, Biochimica et biophysica acta.

[10]  J. Thrasher,et al.  The role of soy phytoestrogens in prostate cancer , 2005, Current opinion in urology.

[11]  Inacio Mandomando,et al.  Efficacy of the RTS,S/AS02A vaccine against Plasmodium falciparum infection and disease in young African children: randomised controlled trial , 2004, The Lancet.

[12]  Pauline Ward,et al.  Protein kinases of the human malaria parasite Plasmodium falciparum: the kinome of a divergent eukaryote , 2004, BMC Genomics.

[13]  Chris J Janse,et al.  A Plasmodium berghei reference line that constitutively expresses GFP at a high level throughout the complete life cycle. , 2004, Molecular and biochemical parasitology.

[14]  J. Kim,et al.  Genistein enhances expression of genes involved in fatty acid catabolism through activation of PPARα , 2004, Molecular and Cellular Endocrinology.

[15]  Simona Corso,et al.  Hepatocyte growth factor and its receptor are required for malaria infection , 2003, Nature Medicine.

[16]  C. R. Garcia,et al.  Interruption of the blood-stage cycle of the malaria parasite, Plasmodium chabaudi, by protein tyrosine kinase inhibitors. , 2003, Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas.

[17]  Anthony Lynch,et al.  Investigations into the concept of a threshold for topoisomerase inhibitor-induced clastogenicity. , 2003, Mutagenesis.

[18]  W. Banz,et al.  Soy isoflavones exert antidiabetic and hypolipidemic effects through the PPAR pathways in obese Zucker rats and murine RAW 264.7 cells. , 2003, The Journal of nutrition.

[19]  J. Boutin,et al.  Peroxisome Proliferator-activated Receptor γ (PPARγ) as a Molecular Target for the Soy Phytoestrogen Genistein* , 2003, The Journal of Biological Chemistry.

[20]  D. Altavilla,et al.  Effects of Genistein and Hormone‐Replacement Therapy on Bone Loss in Early Postmenopausal Women: A Randomized Double‐Blind Placebo‐Controlled Study , 2002, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[21]  Kazuto Ito,et al.  Genistein, a soy isoflavone, induces glutathione peroxidase in the human prostate cancer cell lines LNCaP and PC‐3 , 2002, International journal of cancer.

[22]  M. Tsuji,et al.  Detection of malaria liver-stages in mice infected through the bite of a single Anopheles mosquito using a highly sensitive real-time PCR. , 2001, International journal for parasitology.

[23]  D. Doerge,et al.  Dietary genistein inactivates rat thyroid peroxidase in vivo without an apparent hypothyroid effect. , 2000, Toxicology and applied pharmacology.

[24]  K. Siems,et al.  Antiplasmodial activity of isoflavones from Andira inermis. , 2000, Journal of ethnopharmacology.

[25]  J. Rowley,et al.  Dietary bioflavonoids induce cleavage in the MLL gene and may contribute to infant leukemia. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[26]  P. Rangarajan,et al.  Heme metabolism of Plasmodium is a major antimalarial target. , 2000, Biochemical and biophysical research communications.

[27]  M. Ronis,et al.  An LC-MS method to determine concentrations of isoflavones and their sulfate and glucuronide conjugates in urine. , 1999, Clinica chimica acta; international journal of clinical chemistry.

[28]  A. Dluzewski,et al.  Inhibition of invasion and intraerythrocytic development ofPlasmodium falciparum by kinase inhibitors , 1996, Experientia.

[29]  G. di Perri,et al.  Naturally acquired immunity to Plasmodium falciparum , 1991 .

[30]  M. Akbar,et al.  Genistein, an inhibitor of protein tyrosine kinase, is also a competitive antagonist for P1-purinergic (adenosine) receptor in FRTL-5 thyroid cells. , 1994, Biochemical and biophysical research communications.

[31]  A. Jacquemin-Sablon,et al.  Inhibitory effects of the tyrosine kinase inhibitor genistein on mammalian DNA topoisomerase II. , 1989, Cancer research.

[32]  T. Yoshinari,et al.  Effect of genistein on topoisomerase activity and on the growth of [Val 12]Ha-ras-transformed NIH 3T3 cells. , 1988, Biochemical and biophysical research communications.

[33]  M. Shibuya,et al.  Genistein, a specific inhibitor of tyrosine-specific protein kinases. , 1987, The Journal of biological chemistry.

[34]  K. Setchell,et al.  Assessing risks and benefits of genistein and soy. , 2006, Environmental health perspectives.

[35]  M. Tsuji,et al.  Demonstration of heat-shock protein 70 in the sporozoite stage of malaria parasites , 2004, Parasitology Research.

[36]  J. Boutin,et al.  Peroxisome proliferator-activated receptor gamma (PPARgamma ) as a molecular target for the soy phytoestrogen genistein. , 2003, The Journal of biological chemistry.