Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Choukri Ben Mamoun is active.

Publication


Featured researches published by Choukri Ben Mamoun.


Molecular Microbiology | 2001

Co-ordinated programme of gene expression during asexual intraerythrocytic development of the human malaria parasite Plasmodium falciparum revealed by microarray analysis.

Choukri Ben Mamoun; Ilya Y. Gluzman; Christian Hott; Sandra MacMillan; Aloka S. Amarakone; Dustin L. Anderson; Jane M.-R. Carlton; John B. Dame; Debopam Chakrabarti; Rodger K. Martin; Bernard H. Brownstein; Daniel E. Goldberg

Plasmodium falciparum is a protozoan parasite responsible for the most severe forms of human malaria. All the clinical symptoms and pathological changes seen during human infection are caused by the asexual blood stages of Plasmodium. Within host red blood cells, the parasite undergoes enormous developmental changes during its maturation. In order to analyse the expression of genes during intraerythrocytic development, DNA microarrays were constructed and probed with stage‐specific cDNA. Developmental upregulation of specific mRNAs was found to cluster into functional groups and revealed a co‐ordinated programme of gene expression. Those involved in protein synthesis (ribosomal proteins, translation factors) peaked early in development, followed by those involved in metabolism, most dramatically glycolysis genes. Adhesion/invasion genes were turned on later in the maturation process. At the end of intraerythrocytic development (late schizogony), there was a general shut‐off of gene expression, although a small set of genes, including a number of protein kinases, were turned on at this stage. Nearly all genes showed some regulation over the course of development. A handful of genes remained constant and should be useful for normalizing mRNA levels between stages. These data will facilitate functional analysis of the P. falciparum genome and will help to identify genes with a critical role in parasite progression and multiplication in the human host.


Nucleic Acids Research | 2012

Sequencing of the smallest Apicomplexan genome from the human pathogen Babesia microti

Emmanuel Cornillot; Kamel Hadj-Kaddour; Amina Dassouli; Benjamin Noel; Vincent Ranwez; Benoit Vacherie; Yoann Augagneur; Virginie Bres; Aurelie Duclos; Sylvie Randazzo; B. Carcy; Françoise Debierre-Grockiego; Stephane Delbecq; Karina Moubri-Ménage; Hosam Shams-Eldin; Sahar Usmani-Brown; Frédéric Bringaud; Patrick Wincker; Christian P. Vivarès; Ralph T. Schwarz; Theo Schetters; Peter J. Krause; A. Gorenflot; Vincent Berry; Valérie Barbe; Choukri Ben Mamoun

We have sequenced the genome of the emerging human pathogen Babesia microti and compared it with that of other protozoa. B. microti has the smallest nuclear genome among all Apicomplexan parasites sequenced to date with three chromosomes encoding ∼3500 polypeptides, several of which are species specific. Genome-wide phylogenetic analyses indicate that B. microti is significantly distant from all species of Babesidae and Theileridae and defines a new clade in the phylum Apicomplexa. Furthermore, unlike all other Apicomplexa, its mitochondrial genome is circular. Genome-scale reconstruction of functional networks revealed that B. microti has the minimal metabolic requirement for intraerythrocytic protozoan parasitism. B. microti multigene families differ from those of other protozoa in both the copy number and organization. Two lateral transfer events with significant metabolic implications occurred during the evolution of this parasite. The genomic sequencing of B. microti identified several targets suitable for the development of diagnostic assays and novel therapies for human babesiosis.


Journal of Biological Chemistry | 2003

Genome Expression Analysis in Yeast Reveals Novel Transcriptional Regulation by Inositol and Choline and New Regulatory Functions for Opi1p, Ino2p, and Ino4p

Teresa C. Santiago; Choukri Ben Mamoun

In Saccharomyces cerevisiae, genes encoding phospholipid-synthesizing enzymes are regulated by inositol and choline (IC). The current model suggests that when these precursors become limiting, the transcriptional complex Ino2p-Ino4p activates the expression of these genes, whereas repression requires Opi1p and occurs when IC are available. In this study, microarray-based expression analysis was performed to assess the global transcriptional response to IC in a wild-type strain and in the opi1Δ, ino2Δ, and ino4Δ null mutant strains. Fifty genes were either activated or repressed by IC in the wild-type strain, including three already known IC-repressed genes. We demonstrated that the IC response was not limited to genes involved in membrane biogenesis, but encompassed various metabolic pathways such as biotin synthesis, one-carbon compound metabolism, nitrogen-containing compound transport and degradation, cell wall organization and biogenesis, and acetyl-CoA metabolism. The expression of a large number of IC-regulated genes did not change in the opi1Δ, ino2Δ, and ino4Δ strains, thus implicating new regulatory elements in the IC response. Our studies revealed that Opi1p, Ino2p, and Ino4p have dual regulatory activities, acting in both positive and negative transcriptional regulation of a large number of genes, most of which are not regulated by IC and only a subset of which is involved in membrane biogenesis. These data provide the first global response profile of yeast to IC and reveal novel regulatory mechanisms by these precursors.


Journal of Biological Chemistry | 2005

In Vivo Evidence for the Specificity of Plasmodium falciparum Phosphoethanolamine Methyltransferase and Its Coupling to the Kennedy Pathway

Gabriella Pessi; Jae-Yeon Choi; Jennifer M. Reynolds; Dennis R. Voelker; Choukri Ben Mamoun

Unlike humans and yeast, Plasmodium falciparum, the agent of the most severe form of human malaria, utilizes host serine as a precursor for the synthesis of phosphatidylcholine via a plant-like pathway involving phosphoethanolamine methylation. The monopartite phosphoethanolamine methyltransferase, Pfpmt, plays an important role in the biosynthetic pathway of this major phospholipid by providing the precursor phosphocholine via a three-step S-adenosyl-l-methionine-dependent methylation of phosphoethanolamine. In vitro studies showed that Pfpmt has strong specificity for phosphoethanolamine. However, the in vivo substrate (phosphoethanolamine or phosphatidylethanolamine) is not yet known. We used yeast as a surrogate system to express Pfpmt and provide genetic and biochemical evidence demonstrating the specificity of Pfpmt for phosphoethanolamine in vivo. Wild-type yeast cells, which inherently lack phosphoethanolamine methylation, acquire this activity as a result of expression of Pfpmt. The Pfpmt restores the ability of a yeast mutant pem1Δpem2Δ lacking the phosphatidylethanolamine methyltransferase genes to grow in the absence of choline. Lipid analysis of the Pfpmt-complemented pem1Δpem2Δ strain demonstrates the synthesis of phosphatidylcholine but not the intermediates of phosphatidylethanolamine transmethylation. Complementation of the pem1Δpem2Δ mutant relies on specific methylation of phosphoethanolamine but not phosphatidylethanolamine. Interestingly, a mutation in the yeast choline-phosphate cytidylyltransferase gene abrogates the complementation by Pfpmt thus demonstrating that Pfpmt activity is directly coupled to the Kennedy pathway for the de novo synthesis of phosphatidylcholine.


Eukaryotic Cell | 2008

Purine Salvage Pathways in the Intraerythrocytic Malaria Parasite Plasmodium falciparum

Megan J. Downie; Kiaran Kirk; Choukri Ben Mamoun

Malaria is caused by protozoan parasites of the genus Plasmodium . Five species of Plasmodium cause infection in humans, with the majority of lethal cases caused by Plasmodium falciparum . This species is responsible for more than 500 million clinical cases of malaria each year ([59][1]). In the


BMC Genomics | 2008

A systematic approach to understand the mechanism of action of the bisthiazolium compound T4 on the human malaria parasite, Plasmodium falciparum.

Karine G. Le Roch; Jeffrey R. Johnson; Hugues Ahiboh; Duk Won D. Chung; Jacques Prudhomme; David Plouffe; Kerstin Henson; Yingyao Zhou; William H. Witola; John R. Yates; Choukri Ben Mamoun; Elizabeth A. Winzeler; Henri Vial

BackgroundIn recent years, a major increase in the occurrence of drug resistant falciparum malaria has been reported. Choline analogs, such as the bisthiazolium T4, represent a novel class of compounds with strong potency against drug sensitive and resistant P. falciparum clones. Although T4 and its analogs are presumed to target the parasites lipid metabolism, their exact mechanism of action remains unknown. Here we have employed transcriptome and proteome profiling analyses to characterize the global response of P. falciparum to T4 during the intraerythrocytic cycle of this parasite.ResultsNo significant transcriptional changes were detected immediately after addition of T4 despite the drugs effect on the parasite metabolism. Using the Ontology-based Pattern Identification (OPI) algorithm with an increased T4 incubation time, we demonstrated cell cycle arrest and a general induction of genes involved in gametocytogenesis. Proteomic analysis revealed a significant decrease in the level of the choline/ethanolamine-phosphotransferase (PfCEPT), a key enzyme involved in the final step of synthesis of phosphatidylcholine (PC). This effect was further supported by metabolic studies, which showed a major alteration in the synthesis of PC from choline and ethanolamine by the compound.ConclusionOur studies demonstrate that the bisthiazolium compound T4 inhibits the pathways of synthesis of phosphatidylcholine from choline and ethanolamine in P. falciparum, and provide evidence for post-transcriptional regulations of parasite metabolism in response to external stimuli.


Journal of Biological Chemistry | 2008

Disruption of the Plasmodium falciparum PfPMT Gene Results in a Complete Loss of Phosphatidylcholine Biosynthesis via the Serine-Decarboxylase-Phosphoethanolamine-Methyltransferase Pathway and Severe Growth and Survival Defects

William H. Witola; Kamal El Bissati; Gabriella Pessi; Changan Xie; Paul D. Roepe; Choukri Ben Mamoun

Biochemical studies in the human malaria parasite, Plasmodium falciparum, indicated that in addition to the pathway for synthesis of phosphatidylcholine from choline (CDP-choline pathway), the parasite synthesizes this major membrane phospholipid via an alternative pathway named the serine-decarboxylase-phosphoethanolamine-methyltransferase (SDPM) pathway using host serine and ethanolamine as precursors. However, the role the transmethylation of phosphatidylethanolamine plays in the biosynthesis of phosphatidylcholine and the importance of the SDPM pathway in the parasites growth and survival remain unknown. Here, we provide genetic evidence that knock-out of the PfPMT gene encoding the phosphoethanolamine methyltransferase enzyme completely abrogates the biosynthesis of phosphatidylcholine via the SDPM pathway. Lipid analysis in knock-out parasites revealed that unlike in mammalian and yeast cells, methylation of phosphatidylethanolamine to phosphatidylcholine does not occur in P. falciparum, thus making the SDPM and CDP-choline pathways the only routes for phosphatidylcholine biosynthesis in this organism. Interestingly, loss of PfPMT resulted in significant defects in parasite growth, multiplication, and viability, suggesting that this gene plays an important role in the pathogenesis of intraerythrocytic Plasmodium parasites.


Molecular and Biochemical Parasitology | 2002

Choline transport in Leishmania major promastigotes and its inhibition by choline and phosphocholine analogs

Rachel Zufferey; Choukri Ben Mamoun

Phosphatidylcholine is the most abundant phospholipid in the membranes of the human parasite Leishmania. The metabolic pathways leading to its biosynthesis are likely to play a critical role in parasite development and survival and may offer a good target for antileishmanial chemotherapy. Phosphatidylcholine synthesis via the CDP-choline pathway requires transport of the choline precursor from the host. Here, we report the first characterization of choline transport in this parasite, which is carrier-mediated and exhibits Michaelis-Menten kinetics with an apparent K(m) value of 2.5 microM for choline. This process is Na(+)-independent and requires an intact proton gradient to be fully functional. Choline transport into Leishmania is highly specific for choline and is inhibited by the choline carrier inhibitor hemicholinium-3, the channel blocker quinacrine, the antimalarial aminoquinolines quinine and quinidine, the antileishmanial phosphocholine analogs, miltefosine and edelfosine, and by choline analogs, most of which have antimalarial activities. Most importantly, choline analogs kill the promastigote form of the parasite in vitro in the low micromolar range. These results set the stage for the use of choline analogs in antileishmanial chemotherapy and shed new lights on the mechanism of action of the leishmanicidal phosphocholine analogs.


Eukaryotic Cell | 2004

Candida albicans Csy1p Is a Nutrient Sensor Important for Activation of Amino Acid Uptake and Hyphal Morphogenesis

Elisa Brega; Rachel Zufferey; Choukri Ben Mamoun

ABSTRACT Candida albicans is an important human pathogen that displays a remarkable ability to detect changes in its environment and to respond appropriately by changing its cell morphology and physiology. Serum- and amino acid-based media are known to induce filamentous growth in this organism. However, the mechanism by which amino acids induce filamentation is not yet known. Here, we describe the identification and characterization of the primary amino acid sensor of C. albicans, Csy1. We show that Csy1p plays an important role in amino acid sensing and filamentation. Loss of Csy1p results in a lack of amino acid-mediated activation of amino acid transport and a lack of induction of transcription of specific amino acid permease genes. Furthermore, a csy1Δ/csy1Δ strain, lacking Csy1p, is defective in filamentation and displays altered colony morphology in serum- and amino acid-based media. These data provide the first evidence that C. albicans utilizes the amino acid sensor Csy1p to probe its environment, coordinate its nutritional requirements, and determine its morphological state.


BMC Clinical Pharmacology | 2007

Antimalarial activity of the anticancer and proteasome inhibitor bortezomib and its analog ZL3B

Jennifer M. Reynolds; Kamal El Bissati; Jens Brandenburg; Arthur Günzl; Choukri Ben Mamoun

BackgroundThe high rate of mortality due to malaria and the worldwide distribution of parasite resistance to the commonly used antimalarial drugs chloroquine and pyrimethamine emphasize the urgent need for the development of new antimalarial drugs. An alternative approach to the long and uncertain process of designing and developing new compounds is to identify among the armamentarium of drugs already approved for clinical treatment of various human diseases those that may have strong antimalarial activity.MethodsProteasome inhibitor bortezomib (Velcade™: [(1R)-3-methyl-1-[[(2S)-1-oxo-3-phenyl-2-[(pyrazinylcarbonyl) amino]propyl]amino]butyl] boronic acid), which has been approved for treatment of patients with multiple myeloma, and a second boronate analog Z-Leu-Leu-Leu-B(OH)2 (ZL3B), were tested against four different strains of P. falciparum (3D7, HB3, W2 and Dd2) that are either sensitive or have different levels of resistance to the antimalarial drugs pyrimethamine and chloroquine.ResultsBortezomib and ZL3B are equally effective against drug-sensitive and -resistant parasites and block intraerythrocytic development prior to DNA synthesis, but have no effect on parasite egress or invasion.ConclusionThe identification of bortezomib and its analog as potent antimalarial drugs will set the stage for the advancement of this class of compounds, either alone or in combination therapy, for treatment of malaria, and emphasize the need for large-scale screens to identify new antimalarials within the library of clinically approved compounds.

Collaboration


Dive into the Choukri Ben Mamoun's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dennis R. Voelker

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Rachel Zufferey

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel E. Goldberg

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge