Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2018 May 8;9(3):e02293-17.
doi: 10.1128/mBio.02293-17.

CLAG3 Self-Associates in Malaria Parasites and Quantitatively Determines Nutrient Uptake Channels at the Host Membrane

Affiliations

CLAG3 Self-Associates in Malaria Parasites and Quantitatively Determines Nutrient Uptake Channels at the Host Membrane

Ankit Gupta et al. mBio. .

Erratum in

Abstract

Malaria parasites increase host erythrocyte permeability to ions and nutrients via a broad-selectivity channel known as the plasmodial surface anion channel (PSAC), linked to parasite-encoded CLAG3 and two associated proteins. These proteins lack the multiple transmembrane domains typically present in channel-forming proteins, raising doubts about their precise roles. Using the virulent human Plasmodium falciparum parasite, we report that CLAG3 undergoes self-association and that this protein's expression determines channel phenotype quantitatively. We overcame epigenetic silencing of clag3 paralogs and engineered parasites that express two CLAG3 isoforms simultaneously. Stoichiometric expression of these isoforms yielded intermediate channel phenotypes, in agreement with observed trafficking of both proteins to the host membrane. Coimmunoprecipitation and surface labeling revealed formation of CLAG3 oligomers. In vitro selections applied to these transfectant lines yielded distinct mutants with correlated changes in channel activity. These findings support involvement of the identified oligomers in PSAC formation and parasite nutrient acquisition.IMPORTANCE Malaria parasites are globally important pathogens that evade host immunity by replicating within circulating erythrocytes. To facilitate intracellular growth, these parasites increase erythrocyte nutrient uptake through an unusual ion channel. The parasite CLAG3 protein is a key determinant of this channel, but its lack of homology to known ion channels has raised questions about possible mechanisms. Using a new method that allows simultaneous expression of two different CLAG3 proteins, we identify self-association of CLAG3. The two expressed isoforms faithfully traffic to and insert in the host membrane, while remaining associated with two unrelated parasite proteins. Both the channel phenotypes and molecular changes produced upon selections with a highly specific channel inhibitor are consistent with a multiprotein complex that forms the nutrient pore. These studies support direct involvement of the CLAG3 protein in channel formation and are relevant to antimalarial drug discovery projects targeting parasite nutrient acquisition.

Keywords: Plasmodium falciparum; host-pathogen interactions; integrase; ion channels; malaria; molecular biology; multiprotein complexes; nutrient transport.

PubMed Disclaimer

Figures

FIG 1
FIG 1
Intronic attB as a strategy for rapid introduction of complex gene modifications. (A) Schematic showing transfection approach. The attB was introduced into wild-type KC5 by homologous recombination to produce the C3attB parasite; a second transfection using Bxb1 integrase yielded the TFLC3 clone. The tandem Dd2 and 3D7 CLAG3 isoforms are expressed under the native KC5 clag3h promoter and separated during translation due to the T2A skip peptide. After transfection, the Dd2 isoform carries a C-terminal HA tag, while the 3D7 isoform carries a larger miniSOG-FLAG tag (labeled mSG-F). Selectable markers, human dihydrofolate reductase (hDHFR) and blasticidin-S deaminase (BSD), are shown with orange arrows; relevant restriction sites and fragment sizes are shown. (B) Immunoblotting of total cell lysates from KC5 and C3attB lines, probed with mouse polyclonal anti-CLAG3. WT, wild type. (C) Sorbitol-induced osmotic lysis kinetics for KC5 and C3attB (black and red traces, respectively). (D) Mean ± SEM lysis halftimes for these lines. (E) Osmotic lysis kinetics for indicated parasites with addition of 0, 0.6, and 15 µM ISPA-28 (top to bottom traces in each panel, respectively). Note the dose-dependent inhibition in the Dd2 and C3attB parasites. (F) Mean ± SEM permeability in ISPA-28 dose-response experiments as in panel E for wild-type KC5, 3D7, and Dd2 (red, green, and blue circles, respectively) and the C3attB transfectant parasite (red triangles). Solid lines represent fits to y = {a/[1 + (x/c)]} + {(1 − a)/[1 + (x/b)]}, where a, b, and c are constants. ISPA-28 block in C3attB matches that of Dd2. (G) Southern blotting confirms integration in C3attB and TFLC3 parasite genomes. Hybridization was performed with a mixed probe as marked in panel A. Lanes: 1, wild-type KC5 genome; 2, C3attB genome; 3, TFLC3 genome; 4, pCC1-attB-Dd2sLE plasmid; 5, pLN-attP-Dd2nLE-3D7FL plasmid; an additional larger band (>23 kb) in TFLC3 may reflect undigested concatemers formed by integrase-mediated recombination of attP- and attB-containing plasmids. Please see Fig. S2D for increased exposure to allow detection of bands in lanes 1 and 2. (H and I) Immunoblot assays using indicated epitope tag antibodies. TFLC3 expresses both the Dd2-HA-tagged and the 3D7-miniSOG-FLAG-tagged CLAG3 proteins. S and M represent soluble and membrane-associated fractions, respectively, from Percoll-enriched trophozoite-infected cells.
FIG 2
FIG 2
Coexpression and faithful trafficking of tandem CLAG3 isoforms. (A) Indirect immunofluorescence images showing colocalization of Dd2-HA and 3D7-miniSOG-FLAG CLAG3 isoforms in TFLC3 parasites at the schizont stage and in trophozoites after invasion of new erythrocytes. Bar, 5 µm. (B and C) Immunoblot assays performed using TFLC3 cell lysates and indicated epitope tag antibodies without or with extracellular pronase E treatment prior to cell lysis (− and +, respectively). (D and E) Immunoblot assays using anti-CLAG3 antibody and indicated parasites, without and with pronase E treatment (− and +, respectively). While two cleavage products are detected in TFLC3, the C3attB parent produces a single fragment.
FIG 3
FIG 3
CLAG3 oligomerizes at the host erythrocyte membrane. (A) Coimmunoprecipitation of lysates from indicated parasites using anti-FLAG beads and an anti-HA probe. Lanes show solubilized input (I), final wash (W), and eluate (E). (B) Similar coimmunoprecipitations using anti-HA beads and an anti-FLAG probe. (C) Fraction of CLAG3 in hetero-oligomers, quantified by comparison of the eluate to serial dilutions of the input. The eluate band is more intense than a 100× dilution of the input (10−2I).
FIG 4
FIG 4
CLAG3 merodiploids reveal a stoichiometric contribution to PSAC-mediated nutrient uptake. (A) Schematic showing the minimum distinct combinations of host membrane RhopH complexes in the TFLC3 parasite. CLAG3 is shown as blue ribbon dimers embedded in the host membrane; note the permutations of the two C-terminal epitope tags (gold circles and pink ellipses). RhopH2 and RhopH3 are represented in green. (B) Sorbitol uptake kinetics for indicated parasites with addition of 0, 0.3, 0.6, 1.8, 5, and 15 µM ISPA-28 (top to bottom traces in each panel, respectively). Inhibition in TFLC3 and miniSOG2 is intermediate between those observed for 3D7 and Dd2. (C) Ribbon diagrams showing expression of two CLAG3 isoforms in TFLC3 and miniSOG2. (D) Immunofluorescence images showing trafficking of the 3D7-miniSOG-FLAG CLAG3 isoform in miniSOG2. This protein, expressed under the msp2 promoter, colocalizes with RhopH3 in schizonts and is exported normally in trophozoites after invasion of new erythrocytes. Bar, 5 µm. (E and F) Immunoblot assays using membranes from miniSOG2 probed with anti-FLAG (E) and anti-CLAG3 (F) antibodies. Proteins were separated after cells were incubated without and with pronase E treatment (− and +, respectively). Both CLAG3 proteins expressed in this parasite are integral to the erythrocyte membrane. (G) ISPA-28 dose responses from experiments as in panel B. Symbols represent mean ± SEM for wild-type parasites (3D7, white circles; Dd2, black circles) and merodiploid transfectants (TFLC3, blue triangles; miniSOG2, red triangles).
FIG 5
FIG 5
In vitro selections revert channel phenotypes to those of parental lines. Dose responses for PSAC inhibition by ISPA-28 after selections applied to TFLC3 or miniSOG2 (blue and red symbols, respectively). Circles represent clones generated after treatment with sorbitol, and ISPA-28; triangles reflect clones obtained after cultivation in PGIM with this inhibitor. These clones exhibit potencies that match those of Dd2 or 3D7 (reflected by the solid lines, reproduced from the best fits in Fig. 4G).
FIG 6
FIG 6
Changes in CLAG3 expression account for observed selection phenotypes. (A) Schematic showing genomic changes upon selection of TFLC3 to yield the TFLC3.pgim and TFLC3.sorb clones. These clones loop out one or both transfection plasmids to lose their intermediate channel phenotypes. (B) Schematic showing the three clag3 genes present in the miniSOG2 line; the 3D7 clag3.1 gene is expressed from a distinct genomic site through random piggyBac insertion. Distinguishing features of each allele are tabulated above the ribbon. (C) Mean ± SEM normalized expression of indicated clag3 alleles in the miniSOG2 line before (black bars) and after selection with ISPA-28 and PGIM or sorbitol (red and blue bars, respectively). *, significant changes in gene expression (P < 0.05, one-way ANOVA followed by Dunnett’s multiple-comparison test; replicates from 3 independent trials). (D and E) Immunoblot assays using cell lysates from indicated parasites, probed with anti-FLAG (D) or anti-CLAG3 (E) antibodies. (F) DNA sequence chromatograms showing a C-to-T point mutation that changes glutamine 271 to a stop in the miniSOG2.sorb parasite.

Similar articles

Cited by

References

    1. Maier AG, Cooke BM, Cowman AF, Tilley L. 2009. Malaria parasite proteins that remodel the host erythrocyte. Nat Rev Microbiol 7:341–354. doi:10.1038/nrmicro2110. - DOI - PubMed
    1. Hviid L, Jensen AT. 2015. PfEMP1—a parasite protein family of key importance in Plasmodium falciparum malaria immunity and pathogenesis. Adv Parasitol 88:51–84. doi:10.1016/bs.apar.2015.02.004. - DOI - PubMed
    1. Desai SA. 2012. Ion and nutrient uptake by malaria parasite-infected erythrocytes. Cell Microbiol 14:1003–1009. doi:10.1111/j.1462-5822.2012.01790.x. - DOI - PMC - PubMed
    1. Nguitragool W, Bokhari AA, Pillai AD, Rayavara K, Sharma P, Turpin B, Aravind L, Desai SA. 2011. Malaria parasite clag3 genes determine channel-mediated nutrient uptake by infected red blood cells. Cell 145:665–677. doi:10.1016/j.cell.2011.05.002. - DOI - PMC - PubMed
    1. Pillai AD, Nguitragool W, Lyko B, Dolinta K, Butler MM, Nguyen ST, Peet NP, Bowlin TL, Desai SA. 2012. Solute restriction reveals an essential role for clag3-associated channels in malaria parasite nutrient acquisition. Mol Pharmacol 82:1104–1114. doi:10.1124/mol.112.081224. - DOI - PMC - PubMed

Publication types

MeSH terms

Substances

LinkOut - more resources