ArticlePDF Available

Immunological Dominance of Trypanosoma cruzi Tandem Repeat Proteins

American Society for Microbiology
Infection and Immunity
Authors:
  • PAI Life Sciences Inc.

Abstract and Figures

Proteins with tandem repeat (TR) domains have been found in various protozoan parasites, often acting as targets of B-cell responses. However, the extent of the repeats within Trypanosoma cruzi, the causative agent of Chagas’ disease, has not been examined well. Here, we present a systematic survey of the TR genes found in T. cruzi, in comparison with other organisms. Although the characteristics of TR genes varied from organism to organism, the presence of genes having large TR domains was unique to the trypanosomatids examined, including T. cruzi. Sequence analyses of T. cruzi TR genes revealed their divergency; they do not share such characteristics as sequence similarity or biased cellular location predicted by the presence of a signal sequence or transmembrane domain(s). In contrast, T. cruzi TR proteins seemed to possess significant antigenicity. A number of previously characterized T. cruzi antigens were detected by this computational screening, and several of those antigens contained a large TR domain. Further analyses of the T. cruzi genome demonstrated that previously uncharacterized TR proteins in this organism may also be immunodominant. Taken together, T. cruzi is rich in large TR domain-containing proteins with immunological significance; it is worthwhile further analyzing such characteristics of TR proteins as the copy number and consensus sequence of the repeats to determine whether they might contribute to the biological variability of T. cruzi strains with regard to induced immunological responses, host susceptibility, disease outcomes, and pathogenicity.
Content may be subject to copyright.
INFECTION AND IMMUNITY, Sept. 2008, p. 3967–3974 Vol. 76, No. 9
0019-9567/08/$08.000 doi:10.1128/IAI.00604-08
Copyright © 2008, American Society for Microbiology. All Rights Reserved.
Immunological Dominance of Trypanosoma cruzi Tandem Repeat Proteins
Yasuyuki Goto,
1
* Darrick Carter,
1,2
and Steven G. Reed
1
Infectious Disease Research Institute
1
and Protein Advances Inc.,
2
Seattle, Washington
Received 16 May 2008/Returned for modification 20 June 2008/Accepted 30 June 2008
Proteins with tandem repeat (TR) domains have been found in various protozoan parasites, often acting as
targets of B-cell responses. However, the extent of the repeats within Trypanosoma cruzi, the causative agent of
Chagas’ disease, has not been examined well. Here, we present a systematic survey of the TR genes found in
T.cruzi, in comparison with other organisms. Although the characteristics of TR genes varied from organism
to organism, the presence of genes having large TR domains was unique to the trypanosomatids examined,
including T.cruzi. Sequence analyses of T.cruzi TR genes revealed their divergency; they do not share such
characteristics as sequence similarity or biased cellular location predicted by the presence of a signal sequence
or transmembrane domain(s). In contrast, T.cruzi TR proteins seemed to possess significant antigenicity. A
number of previously characterized T.cruzi antigens were detected by this computational screening, and
several of those antigens contained a large TR domain. Further analyses of the T.cruzi genome demonstrated
that previously uncharacterized TR proteins in this organism may also be immunodominant. Taken together,
T.cruzi is rich in large TR domain-containing proteins with immunological significance; it is worthwhile
further analyzing such characteristics of TR proteins as the copy number and consensus sequence of the
repeats to determine whether they might contribute to the biological variability of T.cruzi strains with regard
to induced immunological responses, host susceptibility, disease outcomes, and pathogenicity.
Chagas’ disease results from infection, generally via contam-
inated blood or the bite of an infected insect, by the protozoan
parasite Trypanosoma cruzi, which can live in a variety of tis-
sues in the mammalian host. According to the World Health
Organization Special Programme for Research and Training in
Tropical Disease Report 2002, the disease is endemic in 18
countries in Central and South America. It is estimated that 16
to 18 million individuals are infected with T.cruzi, with 300,000
new cases per year, and the infection causes 21,000 deaths
annually. Chagas’ disease has an acute phase and a chronic
phase. Manifestations in the acute phase include swelling at
the infection site, fever, and hepatosplenomegaly. Following
an asymptomatic period after the acute phase, an estimated
32% of infected individuals develop chronic Chagas’ disease,
often leading to fatal damage to the heart and digestive tract.
Genes encoding proteins with tandem repeat (TR) domains,
defined here as two or more copies of an amino acid pattern,
have been found in a variety of organisms, from prokaryotes to
higher animals. TRs appear to be diverse and provide regular
arrays of spatial and functional groups (38). They are conspic-
uous in structural and cell surface proteins in some organisms
(38, 60). In contrast, previously characterized T.cruzi TR pro-
teins include trans-sialidase, ribosomal protein, flagellar pro-
tein FRA, and cytoplasmic protein CRA (14, 42, 49), which do
not seem to share functional characteristics. Also, the func-
tions of many T.cruzi TR proteins remain unknown due to a
lack of systematic characterization. Although the functions of
TR proteins are disparate and not confined to a single type of
protein, and a common time of expression or cellular localiza-
tion is not consistently observed, one feature appears to be
shared: they are often potent B-cell antigens. The immunolog-
ical significance of TR proteins during bacterial infections has
been reported (3, 31), and even some cancer antigens to which
patients show antibody responses contain TR domains (41, 47).
In some organisms, having a variety of TRs within a given
protein may play an important role in generating variability in
cell surface immunogens and adhesion molecules, thereby
evading the immune system or enhancing pathogenicity (27,
37, 43, 44, 60). In protozoan parasites, TR proteins often serve
as targets of B-cell responses (39, 54). Antibody responses to
TR proteins have been found in Chagas’ disease (14, 28, 34)
and other parasitic diseases such as leishmaniasis (10, 13) and
malaria (16, 17, 40). However, because the immunological
dominance of TR proteins is not restricted to protozoan par-
asites, systematic analyses of TR genes and proteins are re-
quired to see (i) if T.cruzi has more or fewer TR proteins than
other pathogens or organisms do and (ii) whether these TR
proteins have sequence similarity, a biased cellular location, or
shared immunological recognition motifs. For example, a ge-
nome scale analysis of Saccharomyces cerevisiae has revealed
that most genes containing TRs encode cell wall proteins (60).
In a previous study, we have demonstrated that TR proteins of
Leishmania infantum share immunological dominance (26).
This is the only systematic study of the immunological prop-
erties of protozoan parasite TR proteins, and it still remains
unclear whether other protozoan parasites, including T.cruzi,
possess TR proteins with such characteristics.
Here we performed a computational search for TR genes in
T.cruzi, in comparison with various parasitic protozoan (Leish-
mania major,L.infantum,Trypanosoma brucei,Plasmodium
falciparum,Toxoplasma gondii, and Entamoeba histolytica),
fungal (Candida albicans), bacterial (Salmonella enterica and
Mycobacterium tuberculosis), and human genomes. The analy-
sis revealed no biochemical but immunological characteristics
common in the T.cruzi TR proteins. As an indication of its
* Corresponding author. Mailing address: Infectious Disease Re-
search Institute, 1124 Columbia St., Suite 400, Seattle, WA 98104.
Phone: (206) 330-2519. Fax: (206) 381-3678. E-mail: ygoto@idri.org.
Published ahead of print on 14 July 2008.
3967
detection sensitivity, this computational method captured a
number of previously characterized antigens from T.cruzi sug-
gesting the immunological dominance of TR proteins. To fur-
ther validate the immunological significance of TR proteins
from protozoan parasites, we evaluated the antigenicity of pre-
viously uncharacterized T.cruzi TR proteins. The results dem-
onstrated that immunological recognition was a feature com-
mon to the T.cruzi TR proteins, whereas there were no
apparent similarities or biases in their sequences or predicted
cellular locations.
MATERIALS AND METHODS
Bioinformatic screening of TR genes. We obtained DNA sequence data for P.
falciparum 3D7 CDS (coding sequence) version 2.1.4. (without pseudogenes)
(22), L.major CDS version 5.2 (35), L.infantum CDS version 3.0 (51), and T.
brucei Tb927_CDSs_v4_nopseudo (9) from GeneDB (www.genedb.org) (30);
Trypanosoma cruzi Annotated CDS Release 5.1 (20) from TcruziDB (www
.tcruzidb.org/tcruzidb) (2); T.gondii Annotated CDS Release 4.2 from ToxoDB
(www.toxodb.org/toxo) (21); C.albicans open reading frame coding assembly 21
(36, 58) from The Candida Genome Database (www.candidagenome.org) (5); M.
tuberculosis Release R7 (15) from TubercuList (http://genolist.pasteur.fr/TubercuList/);
S.enterica serovar Typhi CT18 (50); and Homo sapiens (59) Hs36.2 CCDS
nucleotide 20070227 from the NCBI database (www.ncbi.nlm.nih.gov/projects
/CCDS/). Tandem Repeats Finder, a program to locate and display TRs in
DNA sequences, was used for these analyses (http://tandem.bu.edu/trf/trf
.html) (8). The program calculates the score according to selected character-
istics of the TR genes such as the period size of the repeat (i.e., the length of
the repeat unit), the number of copies aligned with the consensus pattern, and
the overall percentage of matches between adjacent copies of a pattern. Most
likely, a high score indicates that the gene possesses a large TR domain. In
this study, the genes were regarded as TR genes if the scores from the
Tandem Repeats Finder analysis were 150 or higher. The cutoff value of 150
is likely to eliminate genes with repeat domains shorter than 75 bp. When
more than one TR domain was found within a gene, only the domain with the
highest score was listed or used for further analyses and protein production.
Analyses of the TR genes of T.cruzi.The biochemical properties of each of the
bioinformatically selected T.cruzi TR proteins were further analyzed virtually for
(i) a protein’s molecular mass, isoelectric point, and hydrophobicity and the
presence of a signal sequence and a transmembrane domain; (ii) its known
antigenicity and/or functions by BLAST searches with both DNA and deduced
amino acid sequences against the NCBI database; and (iii) a mass spectrometry-
evidenced protein expression profile (6), available through the database Tcruz-
iDB. Biochemical characteristics such as average hydrophobicity, isoelectric
point, and molecular weight were calculated with the ProteinMachine software
package from Protein Advances Inc., Seattle, WA. To analyze the entire data-
base, a software interface programmed in C# created protein data files as
comma-separated values for export to Excel. Average hydrophobicity or hydro-
philicity plots of each sequence were determined with a modified Kyte-Doolittle
algorithm with scores ranging from 0.6 (most hydrophilic score possible) to 9.0
(most hydrophobic score possible). T.cruzi TR genes were analyzed for their
specificity for T.cruzi, i.e., whether a homologous gene or protein is found in
Leishmania or other organisms, by blasting the DNA and deduced amino acid
sequences against the NCBI database and GeneDB.
Expression of T.cruzi TR proteins. Partial TR domains containing multiple
repeat units were either PCR amplified or synthesized. Partial TR domains of
Tc00.1047053510827.40 (designated Tc2 in this study), Tc00.1047053511821.179
(Tc3), Tc00.1047053509157.120 (Tc4), and Tc00.1047053508119.200 (Tc6) were
amplified by PCR with T.cruzi total DNA and the following primer sets: Tc2, 5
CAA TTA CAT ATG AGC GCG AGC ACC GCC TGG and 3CAA TTA
AAG CTT CTA GTC GCT CAA CAA CCG CAT G; Tc3, 5CAA TTA CAT
ATG GAG AAC GAG GAG CTG CGT G and 3CAA TTA AAG CTT CTA
CGC ACG AAG CTC CTC CAG; Tc4, 5CAA TTA CAT ATG CCG GAG
ACA GCC TCA GTC and 3CAA TTA AAG CTT CTA CGC GTG ACC GTC
CTC GTC; Tc6, 5CAA TTA CAT ATG GCA ACG GAC GAG TTG and 3
CAA TTA AAG CTT CTA GAG CGC AGT CGC ATC CCT G. Partial TR
domains of Tc00.1047053511557.50 (Tc1), Tc00.1047053510217.10 (Tc8), Tc00.
1047053504019.3 (Tc9), Tc00.1047053506495.40 (Tc10), Tc00.1047053506491.20
(Tc12), Tc00.1047053506559.559 (Tc13), and Tc00.1047053507049.119 (Tc15)
were synthesized by Blue Heron Biotechnology, Inc. (Bothell, WA). The ampli-
fied PCR products or synthesized oligonucleotides were inserted in frame with
the six-His tag of vector pET-28a. The vectors were then transformed into the
Escherichia coli Rosetta strain. The transformed E.coli cells were grown in 2
yeast extract-Tryptone medium, and expression of the recombinant proteins was
induced by cultivation with 1 mM isopropyl--D-thiogalactopyranoside for 3 h.
After lysing cells by sonication and centrifuging them at 10,000 g, the super-
natants were used for purifying the proteins as six-His-tagged proteins with
Ni-nitrilotriacetic acid agarose (Qiagen Inc., Valencia, CA). Proteins were bound
to the resin, washed with sodium deoxycholate-containing buffer, and eluted with
buffer containing 250 M imidazole. The eluted protein was dialyzed against
phosphate-buffered saline (pH 7.4), and the concentration of the purified protein
was measured by the bicinchoninic acid protein assay (Pierce Biotechnology Inc.,
Rockford, IL). The purity of the proteins was assessed by Coomassie blue
staining following sodium dodecyl sulfate-polyacrylamide gel electrophoresis.
Antibody ELISA. The expressed T.cruzi TR proteins were analyzed for sero-
reactivity with sera from Brazilian or Ecuadorian Chagas’ disease patients (n
24). Sera from Brazilian visceral leishmaniasis (VL) patients (n16) and
healthy Brazilian people were used as controls. Proteins were diluted in enzyme-
linked immunosorbent assay (ELISA) coating buffer, and 96-well plates were
coated with 200 ng of individual recombinant antigens, followed by blocking with
phosphate-buffered saline containing 0.05% Tween 20 and 1% bovine serum
albumin. Plates were incubated sequentially with human serum samples (1:200
dilution) and with horseradish peroxidase-conjugated anti-human immunoglob-
ulin G (Rockland Immunochemicals, Inc., Gilbertsville, PA). The plates were
developed with tetramethylbenzidine peroxidase substrate (Kirkegaard & Perry
Laboratories, Gaithersburg, MD) and scanned with a microplate reader at 450
nm (570-nm reference). Three additional recombinant proteins were tested as
controls: T.cruzi sterol 24-c methyltransferase (TcSMT) as a conserved antigen
between Trypanosoma and Leishmania species (24), rK39 as a Leishmania-spe-
cific TR antigen (13), and CRA as a T.cruzi-specific TR antigen (42). Statistical
analyses were performed to compare the reactivity of Chagas’ disease patient
sera to individual antigens with that of VL patients or healthy donors by either
unpaired ttest or Mann-Whitney test based on whether the data sets have a
Gaussian distribution.
RESULTS
Search of TR genes from the T.cruzi genome database. With
the exception of S.enterica, all of the organisms examined in
this study had TR genes comprising 1% of their genomes
(Table 1). A markedly higher prevalence of TR genes was
found in P.falciparum and T.gondii (24.61 and 5.70%, respec-
tively). In contrast, the prevalence of TR genes in the trypano-
somatid parasites was no greater than in E.histolytica,C.al-
bicans,M.tuberculosis, and H.sapiens. TR genes with a score
of 2,000 are likely to have a TR domain 1,000 bp long. The
prevalence of these genes in the whole genomes was higher in
the trypanosomatid and the apicomplexan parasites than in the
other examined pathogens or H.sapiens. The trypanosomatid
parasites, including T.cruzi, had a higher prevalence of TR
genes scoring 2,000 in the whole genomes than the apicom-
plexans, although the apicomplexan parasites, especially P.fal-
ciparum, were richer in total TR genes. The trypanosomatid
parasites seemed to have a preference for such large TR genes
according to a higher prevalence of TR genes, scoring 2,000
in all TR genes, and higher mean and median TR scores in
these species compared with others examined in this study.
The high prevalence of large genes in the trypanosomatid
and the apicomplexan parasites may reflect the high preva-
lence of large TR regions in the genes of those parasites. S.
enterica and M.tuberculosis had average gene sizes smaller
than those of the others examined in this study (Fig. 1). C.
albicans and H.sapiens showed gene size distributions over-
lapping those of the trypanosomatid and the apicomplexans,
up to 10,000 bp. The ratios of genes over 10,000 bp, however,
were much higher in the trypanosomatid and the apicomplexan
3968 GOTO ET AL. INFECT.IMMUN.
parasites (0.75 to 1.95%) than any other species, including C.
albicans and H.sapiens (0.01 to 0.05%).
The period size of the TR refers to the length of the repeat
unit. The distribution of the TR period sizes varied among
these organisms (Fig. 2). TR genes in L.infantum were diver-
gent in their repeat motifs, as their period sizes ranged widely,
from 3 to 498, and no particular period size dominated (i.e.,
constituted more than 10%) in any of the TR genes. A similar
pattern was found in L.major and T.brucei (data not shown).
T.cruzi also had a wide distribution of period sizes, although
the overall distribution was shifted to the left compared to L.
infantum, indicating that T.cruzi TR genes are also divergent.
In contrast, 76.2% of the P.falciparum TR genes had period
sizes of 36 bp and 93.4% had period sizes of 72 bp. Al-
though 10% of the TR genes in C.albicans had a period size
of 51 (single peak, Fig. 2), the period sizes in other TR genes
were widely divergent, as seen in Leishmania and Trypano-
soma. There was a more restricted pattern in the period sizes
of human TR genes, which will be discussed below.
Search for functional similarity in T.cruzi TR proteins. We
examined TR genes in T.cruzi,C.albicans,M.tuberculosis, and
H.sapiens for functional similarities. There were 87 TR genes
identified in C.albicans, 25 (28.7%) of which encoded cell
FIG. 1. Gene size distribution of protozoan parasites and other
organisms. All genes in the genome of were sorted by their sizes with
100-bp intervals.
FIG. 2. Unique patterns in period sizes of TR genes. The yaxis
shows the prevalence of TR genes having a particular period size
among all of the TR genes in the same species.
TABLE 1. Numbers of TR genes in selected pathogens and H.sapiens
Species No. of
genes
No. with TR score of: Total TR genes Genes with TR score of 2,000 Mean TR
score
Median TR
Score
150–499 500–999 1,000–1,999 2,000–4,999 5,000–9,999 10,000 No. % of total
genes No. % of total
genes
% of total
TR genes
L.major 9,218 44 15 16 15 10 3 103 1.12 28 0.30 27.20 2,846 715
L.infantum 8,184 35 9 12 21 7 7 91 1.11 35 0.43 38.50 2,651 1,055
T.brucei 8,161 39 18 21 30 9 5 122 1.49 44 0.54 36.10 2,482 1,122
T.cruzi 19,605 154 48 83 69 3 0 357 1.82 72 0.37 20.20 1,183 717
P.falciparum 5,387 1,153 135 29 7 2 0 1,326 24.61 9 0.17 0.70 356 277
T.gondii 7,793 340 53 34 17 0 0 444 5.70 17 0.22 3.80 481 253
E.histolytica 9,905 180 37 2 0 0 0 219 2.21 0 0.00 0.00 342 273
C.albicans 6,107 72 7 5 3 0 0 87 1.42 3 0.05 3.40 448 226
S.enterica 4,395 5 1 1 0 0 0 7 0.16 0 0.00 0.00 594 479
M.tuberculosis 4,005 43 4 2 0 0 0 49 1.22 0 0.00 0.00 334 230
H.sapiens 17,751 235 141 46 6 1 1 430 2.42 8 0.05 1.90 608 433
a
Number of genes with TR scores of 2,000.
VOL. 76, 2008 T.CRUZI TANDEM REPEAT ANTIGENS 3969
surface proteins such as the agglutinin-like sequence family
(33). C.albicans TR genes with higher scores were more likely
to encode cell surface proteins: 10 of 15 TR genes with a score
of 500 and 7 of 8 TR genes with a score of 1,000 encoded
cell surface proteins (the one exception was polyubiquitin).
Such a high prevalence of TR genes encoding cell surface
proteins has been shown in another fungal species, S.cerevisiae
(60). In M.tuberculosis, 49 genes were identified as containing
TRs. Thirty-five (71.4%) of those were categorized in either
the PE family polymorphic GC-rich repetitive sequence (23
genes, 46.9%) or the PPE family (12 genes, 24.5%), values that
were considerably higher than the prevalence in all M.tuber-
culosis genes (1.6 and 1.7% for the PE family polymorphic
GC-rich repetitive sequence and the PPE family, respectively).
The PE and PPE families, the major source of divergence
between the genomes of M.tuberculosis and M.bovis, which
are otherwise 99% similar, often have multiple copies of
repeat motifs and participate in antigenic variation and intra-
macrophage survival of M.tuberculosis (15, 29, 45, 52). As
described above, 215 (50%) of 430 TR genes of H.sapiens had
period sizes of 84 bp or multiples of 84 bp (164, 252, 368, and
420 bp). It was revealed that most of these (209 genes, 48.6%
of all H.sapiens TR proteins) were zinc finger proteins, one of
the largest families of human proteins, composing 2% of the
human proteome. The zinc finger proteins have the character-
istic, approximately 30-amino-acid-long, zinc finger motifs as
sites of binding to typically DNA, often repeated motifs within
a molecule (7, 59).
In contrast, such functional similarities as seen in C.albi-
cans,M.tuberculosis,orH.sapiens were not evident in TR
proteins of T.cruzi, especially in those with high TR scores. Of
357 TR genes identified from T.cruzi (Table 1), 14 were ap-
parently not full length, as they lacked either a start or a stop
codon, and were not analyzed further. The mean and median
CDS lengths of the remaining 343 T.cruzi TR genes were 2,220
and 1,920 bp, respectively, and were larger than those of the
average for all T.cruzi genes (1,513 and 1,152 bp, respectively
(20). The prevalences of proteins having predicted signal se-
quences or transmembrane domains among these T.cruzi TR
proteins were 27.4 and 31.1%, respectively, and were slightly
higher than those in all T.cruzi proteins (16.0 and 26.4%,
respectively [data obtained from TcruziDB]). Higher preva-
lences of TR proteins with predicted signal sequences (41.3%)
or transmembrane domains (40.1%) were found in those hav-
ing scores of 500, but they were not apparent in those with
scores of 500 (16.6 and 23.8%, respectively). As shown in
Table 2, predicted trans-sialidase, mucins, and mucin-associ-
ated surface proteins, the three largest gene families in T.cruzi
(20), constituted 40% of the T.cruzi TR proteins with a score
of 500. In contrast, the category of TR proteins scoring 500
did not have a high prevalence of any particular family and the
majority was categorized as hypothetical proteins lacking
known functional domains. Unlike TR proteins with lower TR
values (500), those with a 500 score had a higher mean
hydrophilicity compared with that of all T.cruzi proteins (Ta-
ble 2, P0.0001 by Mann-Whitney test).
Immunological dominance of T.cruzi TR. For immunolog-
ical analyses of T.cruzi TR, we focused on the 203 (1.04%)
genes with a score of 500. Because the nearly 20,000 T.cruzi
genes analyzed in this study are from its diploid genome (a
haploid T.cruzi genome is estimated to have 12,000 protein-
coding genes) (20), many of the genes, TR genes included, are
represented twice in the pool of analyzed genes. After consol-
idating TR genes with 70% or greater identity, 106 genes with
different TR domains were identified (the top 20 sequences are
shown in Table 3). Of the 106 genes, 10 encoded previously
characterized antigenic repeat motifs: clone 36, CRA, TcD,
B12, B13, SAPA, FRA, TcLo1.2, TcE, and antigen 38 (1, 14,
18, 28, 32, 34, 42). The remaining 96 genes are previously
uncharacterized as encoding antigens.
To examine whether previously uncharacterized T.cruzi TR
proteins are also antigenic and potentially useful as diagnos-
tics, we cloned and expressed recombinant forms of nine of the
remaining proteins listed in Table 3. Two with homology to
Leishmania proteins were excluded to avoid cross-reactivity
with antibody from leishmaniasis patients. Eight of these de-
tected antibodies in Chagas’ disease patient sera, and the re-
sponses were disease specific; the antibody recognition pat-
terns by these antigens were similar to that of CRA and
contrast with that of TcSMT or rK39 (Fig. 3). Therefore, at
least 13 of the top 20 TR proteins, including the five previously
characterized antigens, are antigenic.
DISCUSSION
TR proteins have been implicated in the ability to influence
host immune responses to protozoan parasites and possibility
to contribute to parasite survival. We previously described the
antigenicity of TRs in Leishmania (26). In this study, we dem-
onstrated that TR proteins from the related trypanosomatid
parasite T.cruzi are also immunodominant, while there is little
or no sequence similarity or apparent bias in cellular location.
These features of TR proteins in the trypanosomatid parasites
contrast with those in C.albicans,M.tuberculosis, and H.sa-
piens, which have biased cellular locations or belong to func-
tional protein families. The immunological dominance of TR
proteins in Leishmania and Trypanosoma parasites is sup-
ported by the fact that antigens of these parasites identified
through serological screening of expression libraries are en-
riched for such proteins relative to the entire proteome. Forty-
TABLE 2. Characteristics of T.cruzi TR proteins
Parameter
No. of proteins
(% of total) with: Total no. of
T.cruzi proteins
(% of total)
c
150–499
TRs
a
500 TRs
b
Presence of:
Signal peptide 62 (41.3) 32 (16.6) 3,141 (16.0)
Transmembrane
domain(s)
61 (40.1) 46 (23.8) 5,169 (26.4)
Gene products:
trans-Sialidase 20 (13.3) 14 (7.3) 735 (3.7)
MASP
d
24 (16.0) 3 (1.6) 938 (4.8)
Mucin 22 (14.7) 2 (1.0) 662 (3.4)
Hypothetical protein 38 (25.3) 131 (67.9) 11,171 (57.0)
a
No. of genes, 150. Mean hydrophobicity score, 4.124.
b
No. of genes, 193. Mean hydrophobicity score, 3.868 (P0.0001 by Mann-
Whitney test compared with hydrophobicity of all T.cruzi proteins).
c
No. of genes, 19,605. Mean hydrophobicity score, 4.177.
d
MASP, mucin-associated surface proteins.
3970 GOTO ET AL. INFECT.IMMUN.
four percent of Leishmania antigens identified by serological
screening of the expression library were TR proteins, whereas
such proteins compose only 1% of the proteome (25, 26).
There are 37 T.cruzi proteins cited as defined serological
antigens in the review article by da Silveira et al. (19), and 9
(24%) of them are TR proteins. When the prevalence of TR
proteins in the T.cruzi proteome is considered (2%), the
likelihood that such proteins are antigenic is significantly
FIG. 3. Antigenic properties of T.cruzi TR proteins. Newly identified TR proteins and previously characterized Leishmania-specific antigen
rK39, T.cruzi-specific antigen CRA, and conserved antigen TcSMT were evaluated by ELISA for reactivity with sera from VL patients (n16),
Chagas’ disease patients (n24), and healthy controls (n8). OD, optical density; ns, not significant; *,P0.05; **,P0.01; ***,P0.001;
****,P0.0001 (by either unpaired ttest or Mann-Whitney test).
TABLE 3. Top 20 TR genes of T.cruzi
Gene
no. Identity
No. of
similar
TR genes
Size
(kDa)
PS
a
(bp) CN
b
TR
score %TR
c
MS
d
expression
Export
property
e
Seroreactivity
f
Leishmania
homolog
g
Name
h
Reference
1 Tc00.1047053510217.10 11 163 195 21.7 7,161 100 No No Unknown No Tc8
2 Tc00.1047053511557.50 3 284 126 36.4 6,969 61 No TM Unknown No Tc1
3 Tc00.1047053504019.3 1 206 42 103.4 6,904 80 T No Unknown No Tc9
4 Tc00.1047053511633.79 1 126 114 22 4,849 77 AEMT SP Clone 36 No 34
5 Tc00.1047053510827.40 0 158 486 7.1 4,802 88 No No Unknown No
6 Tc00.1047053506495.40 3 106 45 65.1 4,613 93 No SP, TM Unknown No Tc10
7 Tc00.1047053506303.80 1 141 126 24.6 4,569 77 No No CRA No 42
8 Tc00.1047053509265.110 4 185 15 178 4,386 51 No TM TcD No 14
9 Tc00.1047053506491.20 2 399 126 71.2 4,384 41 No No Unknown No Tc12
10 Tc00.1047053506559.559 0 NA 72 30.9 4,017 29 M NA
i
Unknown No Tc13
11 Tc00.1047053508831.150 1 171 60 59.1 3,858 71 No No B12 Yes 28
12 Tc00.1047053511671.60 1 125 36 72.4 3,842 69 No No B13 No 28
13 Tc00.1047053511821.179 1 144 105 21.2 3,753 60 AEMT No Unknown No Tc3
14 Tc00.1047053509157.120 1 116 126 15.5 3,555 62 No No Unknown No Tc4
15 Tc00.1047053503617.20 1 218 30 99.9 3,524 51 No No Unknown Yes
16 Tc00.1047053511805.20 1 83 318 5.5 3,477 85 No No Unknown No
17 Tc00.1047053509269.4 1 150 27 111.1 3,392 73 No No Unknown Yes
18 Tc00.1047053506777.110 1 157 117 15.3 3,267 44 A No Unknown No
19 Tc00.1047053508119.200 0 103 117 14.1 3,191 55 No No Unknown No Tc6
20 Tc00.1047053507049.119 1 128 60 30.5 3,160 49 No No Unknown No Tc15
a
PS, period size.
b
CN, copy number.
c
Percentage of TR domains in nucleotide sequence of entire gene.
d
MS, mass spectroscopy-based protein expression evidence. A, amastigote; E, epimastigote; M, metacyclic trypomastigote; T, trypomastigote.
e
Presence of predicted signal sequence (SP) or transmembrane domain(s) (TM).
f
Unknown: serological reactivity not reported before; other entries are the names of previously characterized antigens.
g
Presence of Leishmania proteins with homologous repeat motifs with 60% amino acid sequence identity as the cutoff.
h
Names of recombinant proteins designated for this study.
i
NA, no data available.
VOL. 76, 2008 T.CRUZI TANDEM REPEAT ANTIGENS 3971
higher than that observed for non-TR proteins (P0.0001 by
Fisher’s exact test). The role of such proteins in parasite sur-
vival remains to be defined. A2, one of the Leishmania TR
proteins, was shown to be a virulence factor of L.donovani, the
causative agent of VL; it has more than 40 copies of a 10-
amino-acid repeat, whereas A2 in L.major, which causes cu-
taneous leishmaniasis, has only 1 copy of the repeat, suggesting
that multiple repeats in the A2 protein may play a role in the
visceralization of the parasites (62, 63). We have found other
pairs of TR genes in these Leishmania species; overall se-
quences are highly conserved in both non-TR and TR do-
mains, with the major difference being in the copy number of
the repeat (data not shown), suggesting that the chromosome
bias was set before the divergence of these species or selective
pressure on certain alleles caused the expansion or loss of TR
regions in ancestor genes. Genetically distinct strains of T.cruzi
are responsible for different clinical syndromes in humans (12,
46, 48). For example, the T.cruzi Z12 zymodeme tends to
cause the acute form of Chagas’ disease, whereas the Z1 zy-
modeme preferentially causes chronic disease in humans (48).
It is not known whether variability in TR genes is a primary
factor giving different T.cruzi strains or zymodemes divergent
characteristics relating to clinical outcomes, disease-induced
immune responses, and preference for species of the insect
vector reduviid bugs. Taken together, it is worthwhile analyz-
ing characteristics of TR proteins such as the copy number and
consensus sequence of the repeat(s) to determine whether they
might explain certain biological properties of the parasite and
possibly susceptibility to drugs.
Computational searches for such TR proteins may facilitate
identifying novel antigens from parasite genomes. Identifica-
tion of B-cell antigens from pathogens may facilitate the de-
velopment of diagnostic tests or vaccines. Diagnostic methods
for VL and Chagas’ disease often rely on the detection of
parasite-specific antibodies (53, 56, 57). In Plasmodium,TR
antigens including circumsporozoite protein and merozoite
surface antigen 1 are promising malaria vaccine candidates (23,
55). Traditionally, such targets of B-cell responses have been
identified from parasites through serological screening of an
expression library or by immunoblotting of crude lysate sepa-
rated by two-dimensional gel electrophoresis. In contrast, only
a few attempts have been made to computationally predict
serological antigens of pathogens from the proteome based on
their sequences, such as predicting secreted or surface proteins
(4, 11) and identifying proteins with -helical coiled-coil do-
mains (61). Although the prediction of secreted or surface
proteins has shown some promise in identifying antigens from
T.cruzi (11), it may not be powerful enough to reduce the
number of candidates to a practical level when dealing with the
whole genome. There are 3,141 T.cruzi genes containing se-
quences encoding predicted signal peptides, 5,169 with trans-
membrane domains, and 1,776 containing both. Because a
signal sequence is either present or not, it is impossible to
further prioritize these potentially secreted proteins. There-
fore, new bioinformatic tools to screen sequences for antigen-
encoding genes are needed and searching for TRs may serve as
a powerful method of discovering novel antigens in combina-
tion with other computational methods.
There is a real need for improved diagnostics for Chagas’
disease. T.cruzi parasites are not readily detected during
chronic infection. Therefore, indirect methods are required for
the diagnosis of Chagas’ disease patients and screening for T.
cruzi-contaminated samples. Serological tests for Chagas’ dis-
ease include the indirect fluorescent-antibody test and an
ELISA with whole-cell or recombinant antigens. The Ortho T.
cruzi ELISA Test System (www.orthoclinical.com/chagas
/elisaTestSystem.aspx) is the first such test approved by the
FDA and is used for blood screening. However, serological
tests that use T.cruzi whole-cell lysate have cross-reactivity
with leishmaniasis and other patient sera. This is likely because
a number of proteins are conserved between the related ki-
netoplastid T.cruzi and Leishmania parasites. Leishmania is
endemic in South America, often overlapping in distribution
with T.cruzi. For this reason, we focused on T.cruzi TR
proteins without homology to Leishmania proteins, and such
antigenic proteins will be useful in the development of more
accurate diagnostic tests for Chagas’ disease.
ACKNOWLEDGMENTS
We thank Randy Howard for critical comments and Raodoh Mo-
hamath and Alex Picone for technical assistance.
REFERENCES
1. Affranchino, J. L., C. F. Ibanez, A. O. Luquetti, A. Rassi, M. B. Reyes, R. A.
Macina, L. Aslund, U. Pettersson, and A. C. Frasch. 1989. Identification of
a Trypanosoma cruzi antigen that is shed during the acute phase of Chagas’
disease. Mol. Biochem. Parasitol. 34:221–228.
2. Agu¨ero, F., W. Zheng, D. B. Weatherly, P. Mendes, and J. C. Kissinger. 2006.
TcruziDB: an integrated, post-genomics community resource for Trypano-
soma cruzi. Nucleic Acids Res. 34:D428–431.
3. Allred, D. R., T. C. McGuire, G. H. Palmer, S. R. Leib, T. M. Harkins, T. F.
McElwain, and A. F. Barbet. 1990. Molecular basis for surface antigen size
polymorphisms and conservation of a neutralization-sensitive epitope in
Anaplasma marginale. Proc. Natl. Acad. Sci. USA 87:3220–3224.
4. Ara´oz, R., N. Honore, S. Cho, J. P. Kim, S. N. Cho, M. Monot, C. Demangel,
P. J. Brennan, and S. T. Cole. 2006. Antigen discovery: a postgenomic
approach to leprosy diagnosis. Infect. Immun. 74:175–182.
5. Arnaud, M. B., M. C. Costanzo, M. S. Skrzypek, G. Binkley, C. Lane, S. R.
Miyasato, and G. Sherlock. 2005. The Candida Genome Database (CGD), a
community resource for Candida albicans gene and protein information.
Nucleic Acids Res. 33:D358–D363.
6. Atwood, J. A., III, D. B. Weatherly, T. A. Minning, B. Bundy, C. Cavola, F. R.
Opperdoes, R. Orlando, and R. L. Tarleton. 2005. The Trypanosoma cruzi
proteome. Science 309:473–476.
7. Beerli, R. R., and C. F. Barbas III. 2002. Engineering polydactyl zinc-finger
transcription factors. Nat. Biotechnol. 20:135–141.
8. Benson, G. 1999. Tandem repeats finder: a program to analyze DNA se-
quences. Nucleic Acids Res. 27:573–580.
9. Berriman, M., E. Ghedin, C. Hertz-Fowler, G. Blandin, H. Renauld, D. C.
Bartholomeu, N. J. Lennard, E. Caler, N. E. Hamlin, B. Haas, U. Bohme, L.
Hannick, M. A. Aslett, J. Shallom, L. Marcello, L. Hou, B. Wickstead, U. C.
Alsmark, C. Arrowsmith, R. J. Atkin, A. J. Barron, F. Bringaud, K. Brooks,
M. Carrington, I. Cherevach, T. J. Chillingworth, C. Churcher, L. N. Clark,
C. H. Corton, A. Cronin, R. M. Davies, J. Doggett, A. Djikeng, T. Feldblyum,
M. C. Field, A. Fraser, I. Goodhead, Z. Hance, D. Harper, B. R. Harris, H.
Hauser, J. Hostetler, A. Ivens, K. Jagels, D. Johnson, J. Johnson, K. Jones,
A. X. Kerhornou, H. Koo, N. Larke, S. Landfear, C. Larkin, V. Leech, A.
Line, A. Lord, A. Macleod, P. J. Mooney, S. Moule, D. M. Martin, G. W.
Morgan, K. Mungall, H. Norbertczak, D. Ormond, G. Pai, C. S. Peacock, J.
Peterson, M. A. Quail, E. Rabbinowitsch, M. A. Rajandream, C. Reitter,
S. L. Salzberg, M. Sanders, S. Schobel, S. Sharp, M. Simmonds, A. J.
Simpson, L. Tallon, C. M. Turner, A. Tait, A. R. Tivey, S. Van Aken, D.
Walker, D. Wanless, S. Wang, B. White, O. White, S. Whitehead, J. Wood-
ward, J. Wortman, M. D. Adams, T. M. Embley, K. Gull, E. Ullu, J. D. Barry,
A. H. Fairlamb, F. Opperdoes, B. G. Barrell, J. E. Donelson, N. Hall, C. M.
Fraser, S. E. Melville, and N. M. El-Sayed. 2005. The genome of the African
trypanosome Trypanosoma brucei. Science 309:416–422.
10. Bhatia, A., N. S. Daifalla, S. Jen, R. Badaro, S. G. Reed, and Y. A. Skeiky.
1999. Cloning, characterization and serological evaluation of K9 and K26:
two related hydrophilic antigens of Leishmania chagasi. Mol. Biochem. Para-
sitol. 102:249–261.
11. Bhatia, V., M. Sinha, B. Luxon, and N. Garg. 2004. Utility of the Trypano-
soma cruzi sequence database for identification of potential vaccine candi-
dates by in silico and in vitro screening. Infect. Immun. 72:6245–6254.
3972 GOTO ET AL. INFECT.IMMUN.
12. Brenie`re, S. F., R. Carrasco, S. Revollo, G. Aparicio, P. Desjeux, and M.
Tibayrenc. 1989. Chagas’ disease in Bolivia: clinical and epidemiological
features and zymodeme variability of Trypanosoma cruzi strains isolated
from patients. Am. J. Trop. Med. Hyg. 41:521–529.
13. Burns, J. M., Jr., W. G. Shreffler, D. R. Benson, H. W. Ghalib, R. Badaro,
and S. G. Reed. 1993. Molecular characterization of a kinesin-related antigen
of Leishmania chagasi that detects specific antibody in African and American
visceral leishmaniasis. Proc. Natl. Acad. Sci. USA 90:775–779.
14. Burns, J. M., Jr., W. G. Shreffler, D. E. Rosman, P. R. Sleath, C. J. March,
and S. G. Reed. 1992. Identification and synthesis of a major conserved
antigenic epitope of Trypanosoma cruzi. Proc. Natl. Acad. Sci. USA 89:1239–
1243.
15. Cole, S. T., R. Brosch, J. Parkhill, T. Garnier, C. Churcher, D. Harris, S. V.
Gordon, K. Eiglmeier, S. Gas, C. E. Barry III, F. Tekaia, K. Badcock, D.
Basham, D. Brown, T. Chillingworth, R. Connor, R. Davies, K. Devlin, T.
Feltwell, S. Gentles, N. Hamlin, S. Holroyd, T. Hornsby, K. Jagels, A. Krogh,
J. McLean, S. Moule, L. Murphy, K. Oliver, J. Osborne, M. A. Quail, M. A.
Rajandream, J. Rogers, S. Rutter, K. Seeger, J. Skelton, R. Squares, S.
Squares, J. E. Sulston, K. Taylor, S. Whitehead, and B. G. Barrell. 1998.
Deciphering the biology of Mycobacterium tuberculosis from the complete
genome sequence. Nature 393:537–544.
16. Coppel, R. L., A. F. Cowman, R. F. Anders, A. E. Bianco, R. B. Saint, K. R.
Lingelbach, D. J. Kemp, and G. V. Brown. 1984. Immune sera recognize on
erythrocytes Plasmodium falciparum antigen composed of repeated amino
acid sequences. Nature 310:789–792.
17. Dame, J. B., J. L. Williams, T. F. McCutchan, J. L. Weber, R. A. Wirtz, W. T.
Hockmeyer, W. L. Maloy, J. D. Haynes, I. Schneider, D. Roberts, et al. 1984.
Structure of the gene encoding the immunodominant surface antigen on the
sporozoite of the human malaria parasite Plasmodium falciparum. Science
225:593–599.
18. DaRocha, W. D., D. C. Bartholomeu, C. D. Macedo, M. F. Horta, E. Cunha-
Neto, J. E. Donelson, and S. M. Teixeira. 2002. Characterization of cDNA
clones encoding ribonucleoprotein antigens expressed in Trypanosoma cruzi
amastigotes. Parasitol. Res. 88:292–300.
19. da Silveira, J. F., E. S. Umezawa, and A. O. Luquetti. 2001. Chagas disease:
recombinant Trypanosoma cruzi antigens for serological diagnosis. Trends
Parasitol. 17:286–291.
20. El-Sayed, N. M., P. J. Myler, D. C. Bartholomeu, D. Nilsson, G. Aggarwal,
A. N. Tran, E. Ghedin, E. A. Worthey, A. L. Delcher, G. Blandin, S. J.
Westenberger, E. Caler, G. C. Cerqueira, C. Branche, B. Haas, A. Anupama,
E. Arner, L. Aslund, P. Attipoe, E. Bontempi, F. Bringaud, P. Burton, E.
Cadag, D. A. Campbell, M. Carrington, J. Crabtree, H. Darban, J. F. da
Silveira, P. de Jong, K. Edwards, P. T. Englund, G. Fazelina, T. Feldblyum,
M. Ferella, A. C. Frasch, K. Gull, D. Horn, L. Hou, Y. Huang, E. Kindlund,
M. Klingbeil, S. Kluge, H. Koo, D. Lacerda, M. J. Levin, H. Lorenzi, T.
Louie, C. R. Machado, R. McCulloch, A. McKenna, Y. Mizuno, J. C. Mot-
tram, S. Nelson, S. Ochaya, K. Osoegawa, G. Pai, M. Parsons, M. Pentony,
U. Pettersson, M. Pop, J. L. Ramirez, J. Rinta, L. Robertson, S. L. Salzberg,
D. O. Sanchez, A. Seyler, R. Sharma, J. Shetty, A. J. Simpson, E. Sisk, M. T.
Tammi, R. Tarleton, S. Teixeira, S. Van Aken, C. Vogt, P. N. Ward, B.
Wickstead, J. Wortman, O. White, C. M. Fraser, K. D. Stuart, and B.
Andersson. 2005. The genome sequence of Trypanosoma cruzi, etiologic
agent of Chagas disease. Science 309:409–415.
21. Gajria, B., A. Bahl, J. Brestelli, J. Dommer, S. Fischer, X. Gao, M. Heiges,
J. Iodice, J. C. Kissinger, A. J. Mackey, D. F. Pinney, D. S. Roos, C. J.
Stoeckert, Jr., H. Wang, and B. P. Brunk. 2008. ToxoDB: an integrated
Toxoplasma gondii database resource. Nucleic Acids Res. 36(Database is-
sue):D553–D556.
22. Gardner, M. J., N. Hall, E. Fung, O. White, M. Berriman, R. W. Hyman,
J. M. Carlton, A. Pain, K. E. Nelson, S. Bowman, I. T. Paulsen, K. James,
J. A. Eisen, K. Rutherford, S. L. Salzberg, A. Craig, S. Kyes, M. S. Chan, V.
Nene, S. J. Shallom, B. Suh, J. Peterson, S. Angiuoli, M. Pertea, J. Allen, J.
Selengut, D. Haft, M. W. Mather, A. B. Vaidya, D. M. Martin, A. H. Fair-
lamb, M. J. Fraunholz, D. S. Roos, S. A. Ralph, G. I. McFadden, L. M.
Cummings, G. M. Subramanian, C. Mungall, J. C. Venter, D. J. Carucci,
S. L. Hoffman, C. Newbold, R. W. Davis, C. M. Fraser, and B. Barrell. 2002.
Genome sequence of the human malaria parasite Plasmodium falciparum.
Nature 419:498–511.
23. Girard, M. P., Z. H. Reed, M. Friede, and M. P. Kieny. 2007. A review of
human vaccine research and development: malaria. Vaccine 25:1567–1580.
24. Goto, Y., L. Y. Bogatzki, S. Bertholet, R. N. Coler, and S. G. Reed. 2007.
Protective immunization against visceral leishmaniasis using Leishmania ste-
rol 24-c-methyltransferase formulated in adjuvant. Vaccine 25:7450–7458.
25. Goto, Y., R. N. Coler, J. Guderian, R. Mohamath, and S. G. Reed. 2006.
Cloning, characterization, and serodiagnostic evaluation of Leishmania in-
fantum tandem repeat proteins. Infect. Immun. 74:3939–3945.
26. Goto, Y., R. N. Coler, and S. G. Reed. 2007. Bioinformatic identification of
tandem repeat antigens of the Leishmania donovani complex. Infect. Immun.
75:846–851.
27. Gravekamp, C., B. Rosner, and L. C. Madoff. 1998. Deletion of repeats in the
alpha C protein enhances the pathogenicity of group B streptococci in
immune mice. Infect. Immun. 66:4347–4354.
28. Gruber, A., and B. Zingales. 1993. Trypanosoma cruzi: characterization of
two recombinant antigens with potential application in the diagnosis of
Chagas’ disease. Exp. Parasitol. 76:1–12.
29. Hermans, P. W., D. van Soolingen, and J. D. van Embden. 1992. Character-
ization of a major polymorphic tandem repeat in Mycobacterium tuberculosis
and its potential use in the epidemiology of Mycobacterium kansasii and
Mycobacterium gordonae. J. Bacteriol. 174:4157–4165.
30. Hertz-Fowler, C., C. S. Peacock, V. Wood, M. Aslett, A. Kerhornou, P.
Mooney, A. Tivey, M. Berriman, N. Hall, K. Rutherford, J. Parkhill, A. C.
Ivens, M. A. Rajandream, and B. Barrell. 2004. GeneDB: a resource for
prokaryotic and eukaryotic organisms. Nucleic Acids Res. 32:D339–D343.
31. Hollingshead, S. K., V. A. Fischetti, and J. R. Scott. 1986. Complete nucle-
otide sequence of type 6 M protein of the group A streptococcus. Repetitive
structure and membrane anchor. J. Biol. Chem. 261:1677–1686.
32. Houghton, R. L., D. R. Benson, L. D. Reynolds, P. D. McNeill, P. R. Sleath,
M. J. Lodes, Y. A. Skeiky, D. A. Leiby, R. Badaro, and S. G. Reed. 1999. A
multi-epitope synthetic peptide and recombinant protein for the detection of
antibodies to Trypanosoma cruzi in radioimmunoprecipitation-confirmed
and consensus-positive sera. J. Infect. Dis. 179:1226–1234.
33. Hoyer, L. L., C. B. Green, S. H. Oh, and X. Zhao. 2008. Discovering the
secrets of the Candida albicans agglutinin-like sequence (ALS) gene family—a
sticky pursuit. Med. Mycol. 46:1–15.
34. Iban˜ez, C. F., J. L. Affranchino, R. A. Macina, M. B. Reyes, S. Leguizamon,
M. E. Camargo, L. Aslund, U. Pettersson, and A. C. Frasch. 1988. Multiple
Trypanosoma cruzi antigens containing tandemly repeated amino acid se-
quence motifs. Mol. Biochem. Parasitol. 30:27–33.
35. Ivens, A. C., C. S. Peacock, E. A. Worthey, L. Murphy, G. Aggarwal, M.
Berriman, E. Sisk, M. A. Rajandream, E. Adlem, R. Aert, A. Anupama, Z.
Apostolou, P. Attipoe, N. Bason, C. Bauser, A. Beck, S. M. Beverley, G.
Bianchettin, K. Borzym, G. Bothe, C. V. Bruschi, M. Collins, E. Cadag, L.
Ciarloni, C. Clayton, R. M. Coulson, A. Cronin, A. K. Cruz, R. M. Davies, J.
De Gaudenzi, D. E. Dobson, A. Duesterhoeft, G. Fazelina, N. Fosker, A. C.
Frasch, A. Fraser, M. Fuchs, C. Gabel, A. Goble, A. Goffeau, D. Harris, C.
Hertz-Fowler, H. Hilbert, D. Horn, Y. Huang, S. Klages, A. Knights, M.
Kube, N. Larke, L. Litvin, A. Lord, T. Louie, M. Marra, D. Masuy, K.
Matthews, S. Michaeli, J. C. Mottram, S. Muller-Auer, H. Munden, S.
Nelson, H. Norbertczak, K. Oliver, S. O’Neil, M. Pentony, T. M. Pohl, C.
Price, B. Purnelle, M. A. Quail, E. Rabbinowitsch, R. Reinhardt, M. Rieger,
J. Rinta, J. Robben, L. Robertson, J. C. Ruiz, S. Rutter, D. Saunders, M.
Schafer, J. Schein, D. C. Schwartz, K. Seeger, A. Seyler, S. Sharp, H. Shin,
D. Sivam, R. Squares, S. Squares, V. Tosato, C. Vogt, G. Volckaert, R.
Wambutt, T. Warren, H. Wedler, J. Woodward, S. Zhou, W. Zimmermann,
D. F. Smith, J. M. Blackwell, K. D. Stuart, B. Barrell, and P. J. Myler. 2005.
The genome of the kinetoplastid parasite, Leishmania major. Science 309:
436–442.
36. Jones, T., N. A. Federspiel, H. Chibana, J. Dungan, S. Kalman, B. B. Magee,
G. Newport, Y. R. Thorstenson, N. Agabian, P. T. Magee, R. W. Davis, and
S. Scherer. 2004. The diploid genome sequence of Candida albicans. Proc.
Natl. Acad. Sci. USA 101:7329–7334.
37. Jordan, P., L. A. Snyder, and N. J. Saunders. 2003. Diversity in coding
tandem repeats in related Neisseria spp. BMC Microbiol. 3:23.
38. Katti, M. V., R. Sami-Subbu, P. K. Ranjekar, and V. S. Gupta. 2000. Amino
acid repeat patterns in protein sequences: their diversity and structural-
functional implications. Protein Sci. 9:1203–1209.
39. Kemp, D. J., R. L. Coppel, and R. F. Anders. 1987. Repetitive proteins and
genes of malaria. Annu. Rev. Microbiol. 41:181–208.
40. Koenen, M., A. Scherf, O. Mercereau, G. Langsley, L. Sibilli, P. Dubois, L.
Pereira da Silva, and B. Muller-Hill. 1984. Human antisera detect a Plas-
modium falciparum genomic clone encoding a nonapeptide repeat. Nature
311:382–385.
41. Kotera, Y., J. D. Fontenot, G. Pecher, R. S. Metzgar, and O. J. Finn. 1994.
Humoral immunity against a tandem repeat epitope of human mucin
MUC-1 in sera from breast, pancreatic, and colon cancer patients. Cancer
Res. 54:2856–2860.
42. Lafaille, J. J., J. Linss, M. A. Krieger, T. Souto-Padron, W. de Souza, and S.
Goldenberg. 1989. Structure and expression of two Trypanosoma cruzi genes
encoding antigenic proteins bearing repetitive epitopes. Mol. Biochem. Para-
sitol. 35:127–136.
43. Levdansky, E., J. Romano, Y. Shadkchan, H. Sharon, K. J. Verstrepen, G. R.
Fink, and N. Osherov. 2007. Coding tandem repeats generate diversity in
Aspergillus fumigatus genes. Eukaryot. Cell 6:1380–1391.
44. Madoff, L. C., J. L. Michel, E. W. Gong, D. E. Kling, and D. L. Kasper. 1996.
Group B streptococci escape host immunity by deletion of tandem repeat
elements of the alpha C protein. Proc. Natl. Acad. Sci. USA 93:4131–4136.
45. Marri, P. R., J. P. Bannantine, and G. B. Golding. 2006. Comparative
genomics of metabolic pathways in Mycobacterium species: gene duplica-
tion, gene decay and lateral gene transfer. FEMS Microbiol. Rev. 30:906–
925.
46. Miles, M. A., R. A. Cedillos, M. M. Povoa, A. A. de Souza, A. Prata, and V.
Macedo. 1981. Do radically dissimilar Trypanosoma cruzi strains (zymo-
demes) cause Venezuelan and Brazilian forms of Chagas’ disease? Lancet
1:1338–1340.
VOL. 76, 2008 T.CRUZI TANDEM REPEAT ANTIGENS 3973
47. Mollick, J. A., F. S. Hodi, R. J. Soiffer, L. M. Nadler, and G. Dranoff. 2003.
MUC1-like tandem repeat proteins are broadly immunogenic in cancer
patients. Cancer Immun. 3:3.
48. Montamat, E. E., G. M. De Luca D’Oro, R. H. Gallerano, R. Sosa, and A.
Blanco. 1996. Characterization of Trypanosoma cruzi populations by zymo-
demes: correlation with clinical picture. Am. J. Trop. Med. Hyg. 55:625–628.
49. Pais, F. S., W. D. Darocha, R. M. Almeida, S. Y. Leclercq, M. L. Penido, S. P.
Fragoso, D. C. Bartholomeu, R. T. Gazzinelli, and S. M. Teixeira. 2008.
Molecular characterization of ribonucleoproteic antigens containing re-
peated amino acid sequences from Trypanosoma cruzi. Microbes Infect.
10:716–725.
50. Parkhill, J., G. Dougan, K. D. James, N. R. Thomson, D. Pickard, J. Wain,
C. Churcher, K. L. Mungall, S. D. Bentley, M. T. Holden, M. Sebaihia, S.
Baker, D. Basham, K. Brooks, T. Chillingworth, P. Connerton, A. Cronin, P.
Davis, R. M. Davies, L. Dowd, N. White, J. Farrar, T. Feltwell, N. Hamlin,
A. Haque, T. T. Hien, S. Holroyd, K. Jagels, A. Krogh, T. S. Larsen, S.
Leather, S. Moule, P. O’Gaora, C. Parry, M. Quail, K. Rutherford, M.
Simmonds, J. Skelton, K. Stevens, S. Whitehead, and B. G. Barrell. 2001.
Complete genome sequence of a multiple drug resistant Salmonella enterica
serovar Typhi CT18. Nature 413:848–852.
51. Peacock, C. S., K. Seeger, D. Harris, L. Murphy, J. C. Ruiz, M. A. Quail, N.
Peters, E. Adlem, A. Tivey, M. Aslett, A. Kerhornou, A. Ivens, A. Fraser,
M. A. Rajandream, T. Carver, H. Norbertczak, T. Chillingworth, Z. Hance,
K. Jagels, S. Moule, D. Ormond, S. Rutter, R. Squares, S. Whitehead, E.
Rabbinowitsch, C. Arrowsmith, B. White, S. Thurston, F. Bringaud, S. L.
Baldauf, A. Faulconbridge, D. Jeffares, D. P. Depledge, S. O. Oyola, J. D.
Hilley, L. O. Brito, L. R. Tosi, B. Barrell, A. K. Cruz, J. C. Mottram, D. F.
Smith, and M. Berriman. 2007. Comparative genomic analysis of three
Leishmania species that cause diverse human disease. Nat. Genet. 39:839–
847.
52. Poulet, S., and S. T. Cole. 1995. Characterization of the highly abundant
polymorphic GC-rich-repetitive sequence (PGRS) present in Mycobacte-
rium tuberculosis. Arch. Microbiol. 163:87–95.
53. Reed, S. G. 1996. Diagnosis of leishmaniasis. Clin. Dermatol. 14:471–478.
54. Reeder, J. C., and G. V. Brown. 1996. Antigenic variation and immune
evasion in Plasmodium falciparum malaria. Immunol. Cell Biol. 74:546–554.
55. Richie, T. L., and A. Saul. 2002. Progress and challenges for malaria vac-
cines. Nature 415:694–701.
56. Singh, S., and R. Sivakumar. 2003. Recent advances in the diagnosis of
leishmaniasis. J. Postgrad. Med. 49:55–60.
57. Sundar, S., and M. Rai. 2002. Laboratory diagnosis of visceral leishmaniasis.
Clin. Diagn. Lab. Immunol. 9:951–958.
58. van het Hoog, M., T. J. Rast, M. Martchenko, S. Grindle, D. Dignard, H.
Hogues, C. Cuomo, M. Berriman, S. Scherer, B. B. Magee, M. Whiteway, H.
Chibana, A. Nantel, and P. T. Magee. 2007. Assembly of the Candida albi-
cans genome into sixteen supercontigs aligned on the eight chromosomes.
Genome. Biol. 8:R52.
59. Venter, J. C., M. D. Adams, E. W. Myers, P. W. Li, R. J. Mural, G. G. Sutton,
H. O. Smith, M. Yandell, C. A. Evans, R. A. Holt, J. D. Gocayne, P. Amanati-
des, R. M. Ballew, D. H. Huson, J. R. Wortman, Q. Zhang, C. D. Kodira,
X. H. Zheng, L. Chen, M. Skupski, G. Subramanian, P. D. Thomas, J.
Zhang, G. L. Gabor Miklos, C. Nelson, S. Broder, A. G. Clark, J. Nadeau,
V. A. McKusick, N. Zinder, A. J. Levine, R. J. Roberts, M. Simon, C.
Slayman, M. Hunkapiller, R. Bolanos, A. Delcher, I. Dew, D. Fasulo, M.
Flanigan, L. Florea, A. Halpern, S. Hannenhalli, S. Kravitz, S. Levy, C.
Mobarry, K. Reinert, K. Remington, J. Abu-Threideh, E. Beasley, K. Bid-
dick, V. Bonazzi, R. Brandon, M. Cargill, I. Chandramouliswaran, R. Char-
lab, K. Chaturvedi, Z. Deng, V. Di Francesco, P. Dunn, K. Eilbeck, C.
Evangelista, A. E. Gabrielian, W. Gan, W. Ge, F. Gong, Z. Gu, P. Guan, T. J.
Heiman, M. E. Higgins, R. R. Ji, Z. Ke, K. A. Ketchum, Z. Lai, Y. Lei, Z. Li,
J. Li, Y. Liang, X. Lin, F. Lu, G. V. Merkulov, N. Milshina, H. M. Moore,
A. K. Naik, V. A. Narayan, B. Neelam, D. Nusskern, D. B. Rusch, S. Salzberg,
W. Shao, B. Shue, J. Sun, Z. Wang, A. Wang, X. Wang, J. Wang, M. Wei, R.
Wides, C. Xiao, C. Yan, A. Yao, J. Ye, M. Zhan, W. Zhang, H. Zhang, Q.
Zhao, L. Zheng, F. Zhong, W. Zhong, S. Zhu, S. Zhao, D. Gilbert, S.
Baumhueter, G. Spier, C. Carter, A. Cravchik, T. Woodage, F. Ali, H. An, A.
Awe, D. Baldwin, H. Baden, M. Barnstead, I. Barrow, K. Beeson, D. Busam,
A. Carver, A. Center, M. L. Cheng, L. Curry, S. Danaher, L. Davenport, R.
Desilets, S. Dietz, K. Dodson, L. Doup, S. Ferriera, N. Garg, A. Gluecks-
mann, B. Hart, J. Haynes, C. Haynes, C. Heiner, S. Hladun, D. Hostin, J.
Houck, T. Howland, C. Ibegwam, J. Johnson, F. Kalush, L. Kline, S. Koduru,
A. Love, F. Mann, D. May, S. McCawley, T. McIntosh, I. McMullen, M. Moy,
L. Moy, B. Murphy, K. Nelson, C. Pfannkoch, E. Pratts, V. Puri, H. Qureshi,
M. Reardon, R. Rodriguez, Y. H. Rogers, D. Romblad, B. Ruhfel, R. Scott, C.
Sitter, M. Smallwood, E. Stewart, R. Strong, E. Suh, R. Thomas, N. N. Tint,
S. Tse, C. Vech, G. Wang, J. Wetter, S. Williams, M. Williams, S. Windsor,
E. Winn-Deen, K. Wolfe, J. Zaveri, K. Zaveri, J. F. Abril, R. Guigo, M. J.
Campbell, K. V. Sjolander, B. Karlak, A. Kejariwal, H. Mi, B. Lazareva, T.
Hatton, A. Narechania, K. Diemer, A. Muruganujan, N. Guo, S. Sato, V.
Bafna, S. Istrail, R. Lippert, R. Schwartz, B. Walenz, S. Yooseph, D. Allen,
A. Basu, J. Baxendale, L. Blick, M. Caminha, J. Carnes-Stine, P. Caulk,
Y. H. Chiang, M. Coyne, C. Dahlke, A. Mays, M. Dombroski, M. Donnelly,
D. Ely, S. Esparham, C. Fosler, H. Gire, S. Glanowski, K. Glasser, A.
Glodek, M. Gorokhov, K. Graham, B. Gropman, M. Harris, J. Heil, S.
Henderson, J. Hoover, D. Jennings, C. Jordan, J. Jordan, J. Kasha, L.
Kagan, C. Kraft, A. Levitsky, M. Lewis, X. Liu, J. Lopez, D. Ma, W. Majoros,
J. McDaniel, S. Murphy, M. Newman, T. Nguyen, N. Nguyen, M. Nodell, S.
Pan, J. Peck, M. Peterson, W. Rowe, R. Sanders, J. Scott, M. Simpson, T.
Smith, A. Sprague, T. Stockwell, R. Turner, E. Venter, M. Wang, M. Wen, D.
Wu, M. Wu, A. Xia, A. Zandieh, and X. Zhu. 2001. The sequence of the
human genome. Science 291:1304–1351.
60. Verstrepen, K. J., A. Jansen, F. Lewitter, and G. R. Fink. 2005. Intragenic
tandem repeats generate functional variability. Nat. Genet. 37:986–990.
61. Villard, V., G. W. Agak, G. Frank, A. Jafarshad, C. Servis, I. Nebie, S. B.
Sirima, I. Felger, M. Arevalo-Herrera, S. Herrera, F. Heitz, V. Backer, P.
Druilhe, A. V. Kajava, and G. Corradin. 2007. Rapid identification of ma-
laria vaccine candidates based on alpha-helical coiled coil protein motif.
PLoS ONE 2:e645.
62. Zhang, W. W., and G. Matlashewski. 1997. Loss of virulence in Leishmania
donovani deficient in an amastigote-specific protein, A2. Proc. Natl. Acad.
Sci. USA 94:8807–8811.
63. Zhang, W. W., S. Mendez, A. Ghosh, P. Myler, A. Ivens, J. Clos, D. L. Sacks,
and G. Matlashewski. 2003. Comparison of the A2 gene locus in Leishmania
donovani and Leishmania major and its control over cutaneous infection.
J. Biol. Chem. 278:35508–35515.
Editor: W. A. Petri, Jr.
3974 GOTO ET AL. INFECT.IMMUN.
... Antigens containing TRs often serve as targets of B-cell responses [11]. In addition, TR antigens have been found in a variety of parasites, including Fasciola hepatica, Leishmania donovani, Plasmodium falciparum and Trypanosoma cruzi [16,[34][35][36]. Regarding C. sinensis, some currently validated antigens are also TR proteins, such as Cs31 [37], CsPRA [38], CsGRP [39] and Cs1 [11], and they are genus-or species-specific [40]. ...
Article
Full-text available
Clonorchis sinensis is one of the most important fish-borne zoonotic parasitic worms in humans, and is distributed in several countries with more than 15 million people infected globally. However, the lack of a point-of-care testing (POCT) method is still the critical barrier to effectively prevent clonorchiasis. With the application of novel fluorescent nanomaterials, the development of on-site testing methods with high signal enhancement can provide a simple, precise and inexpensive tool for disease detection. In this study, Eu-(III) nanoparticles (EuNPs) were used as indicative probes, combined with C . sinensis tandem repeat sequence 1 (CSTR1) antigen to capture specific antibodies. Afterward, the complex binds to mouse anti-human IgG immobilized on the test line (T-line) producing a fluorescent signal under UV light. The EuNPs-fluorescent immunoassay (EuNPs-FIA) was successfully constructed, allowing sample detection within 10 min. It enabled both qualitative determination with the naked eye under UV light and quantitative detection by scanning the fluorescence intensity on the test line and control line (C-line). A total of 133 clinical human sera (74 negative, 59 clonorchiasis, confirmed by conventional Kato-Katz (KK) methods and PCR via testing fecal samples corresponding to each serum sample) were used in this study. For qualitative analysis, the cut-off value of fluorescence for positive serum was 31.57 by testing 74 known negative human samples. The assay had no cross-reaction with other 9 parasite-infected sera, and could recognize the mixed infection sera of C . sinensis and other parasites. The sensitivity and specificity of EuNPs-FIA were both 100% compared with KK smear method. Taking advantage of its high precision and user-friendly procedure, the established EuNPs-FIA provides a powerful tool for the diagnosis and epidemiological survey of clonorchiasis.
... The main targets of the CD8 + T cell response are the parasite antigens exposed at the time of host cell rupture, which mainly include proteins from the trans-sialidases family that cover the surface of T. cruzi and are also secreted. Although trans-sialidase molecules are not the only targets of T. cruzi-specific CD8 + T cells, there appears to be an immunodominance for these epitopes [82][83][84][85]. This immunodominance by trans-sialidases has some drawbacks: TSs have a high variability between strains, based on sequence and expression patterns, and their expression occurs late in the intracellular infection cycle. ...
Article
Full-text available
The neglected Chagas disease (CD) is caused by the protozoan parasite Trypanosoma cruzi. Despite CD dispersion throughout the world, it prevails in tropical areas affecting mainly poor communities, causing devastating health, social and economic consequences. Clinically, CD is marked by a mildly symptomatic acute phase, and a chronic phase characterized by cardiac and/or digestive complications. Current treatment for CD relies on medications with strong side effects and reduced effectiveness. The complex interaction between the parasite and the host outlines the etiology and progression of CD. The unique characteristics and high adaptability of T. cruzi, its mechanisms of persistence, and evasion of the immune system seem to influence the course of the disease. Despite the efforts to uncover the pathology of CD, there are many gaps in understanding how it is established and reaches chronicity. Also, the lack of effective treatments and protective vaccines constitute challenges for public health. Here, we explain the background in which CD is established, from the peculiarities of T. cruzi molecular biology to the development of the host’s immune response leading to the pathophysiology of CD. We also discuss the state of the art of treatments for CD and current challenges in basic and applied science.
... Approximately 25% of the proteins expressed by T. cruzi contain tandem repeat amino acid sequences consisting of 5-68 amino acids [136][137][138][139]. This improved recognition by antibodies compared to proteins that lack repeated sequences [130,140,141] and improved the performance of immunoassays compared to cell extracts or whole epimastigotes [136,142]. Indeed, sera from infected humans often contain high titers of antibodies against these repeated sequences [143][144][145]. ...
Article
Full-text available
Background: Chagas disease (CD) is caused by Trypanosoma cruzi, which is transmitted mainly through the feces/urine of infected triatomine bugs. The acute phase lasts 2–3 months and is characterized by high parasitemia and nonspecific symptoms, whereas the lifelong chronic phase features symptoms affecting the heart and/or digestive tract occurring in 30–40% of infected individuals. As in humans, cardiac abnormalities are observed in T. cruzi-infected dogs and cats. We reviewed the technological advances in the serological diagnosis of CD in dogs and cats. Methods: A review of the published literature during the last 54 years (1968–2022) on the epidemiology, clinical features, diagnosis, treatment and prevention of CD in dogs and cats was conducted. Results: Using predefined eligibility criteria for a search of the published literature, we retrieved and screened 436 publications. Of these, 84 original studies were considered for inclusion in this review. Dogs and cats are considered as sentinels, potentially indicating an active T. cruzi transmission and thus the risk for human infection. Although dogs and cats are reputed to be important for maintaining the T. cruzi domestic transmission cycle, there are no commercial tests to detect past or active infections in these animals. Most published research on CD in dogs and cats have used in-house serological tests prepared with native and/or full-length recombinant antigens, resulting in variable diagnostic performance. In recent years, chimeric antigens have been used to improve the diagnosis of chronic CD in humans with encouraging results. Some of them have high performance values (> 95%) and extremely low crossreactivity rates for Leishmania spp., especially the antigens IBMP-8.1 to IBMP-8.4. The diagnostic performance of IBMP antigens was also investigated in dogs, showing high diagnostic performance with negligible cross-reactivity with anti-Leishmania infantum antibodies. Conclusions: The development of a commercial immunodiagnostic tool to identify past or active T. cruzi infections in dogs and cats is urgently needed. The use of chimeric recombinant T. cruzi antigens may help to fill this gap and is discussed in this review.
... In that sense, it is remarkable that NSG1 is predicted to have linear B cell epitopes throughout its whole sequence, with high scores for hydrophilicity, accessibility, and flexibility exactly in the region where one of the sets of the tandem repeats are concentrated (Fig. S1B). These features have been well characterized for that purpose in other protozoan parasites such as Leishmania and T. cruzi (46,47). ...
Article
Full-text available
Neospora caninum is a parasite with veterinary relevance, inducing severe disease in dogs and reproductive disorders in ruminants, especially cattle, leading to major losses. The close phylogenetic relationship to Toxoplasma gondii and the lack of pathogenicity in humans drives an interest of the scientific community toward using N. caninum as a model to study the pathogenicity of T. gondii .
... These results suggest that the rKDDR-plus antigen is probably capable of interacting with lymphocytes of the host's immune system. These epitopes can be more easily identified during antibody serological screening compared to molecules with fewer repetitions, thus compensating for the low prevalence of antibodies observed in some patients [53]. To evaluate the influence of the number of 39 amino acid motif repetitions from kinesinderived proteins, we performed comparative ELISA assays with the three proteins. ...
Article
Full-text available
Visceral leishmaniasis (VL) is caused by protozoa belonging to the Leishmania donovani complex and is considered the most serious and fatal form among the different types of leishmaniasis, if not early diagnosed and treated. Among the measures of disease control stand out the management of infected dogs and the early diagnosis and appropriate treatment of human cases. Several antigens have been characterized for use in the VL diagnosis, among them are the recombinant kinesin-derived antigens from L . infantum , as rK39 and rKDDR. The main difference between these antigens is the size of the non-repetitive kinesin region and the number of repetitions of the 39 amino acid degenerate motif (6.5 and 8.5 repeats in rK39 and rKDDR, respectively). This repetitive region has a high antigenicity score. To evaluate the effect of increasing the number of repeats on diagnostic performance, we designed the rKDDR-plus antigen, containing 15.3 repeats of the 39 amino acid degenerate motif, besides the absence of the non-repetitive portion from L . infantum kinesin. Its performance was evaluated by enzyme-linked immunosorbent assay (ELISA) and rapid immunochromatographic test (ICT), and compared with the kinesin-derived antigens (rKDDR and rK39). In ELISA with human sera, all recombinant antigens had a sensitivity of 98%, whereas the specificity for rKDDR-plus, rKDDR and rK39 was 100%, 96% and 71%, respectively. When evaluated canine sera, the ELISA sensitivity was 97% for all antigens, and the specificity for rKDDR-plus, rKDDR and rK39 was 98%, 91% and 83%, respectively. Evaluation of the ICT/rKDDR-plus, using human sera, showed greater diagnostic sensitivity (90%) and specificity (100%), when compared to the IT LEISH (79% and 98%, respectively), which is based on the rK39 antigen. These results suggest that the increased presence of repetitive motifs in the rKDDR-plus protein improves the diagnostic performance of serological tests by increasing the specificity and accuracy of the diagnosis.
... In the T. cruzi parasite, a significant number of proteins containing large tandem repeat domains have been shown to have significant immunological relevance, since most of them are antigenic molecules (12). The antigens bearing amino acid tandem repeats seem to possess a significant degree of antigenicity and to be targets of B-cell responses. ...
... In this context, pathogen-specific T cells appear to preferentially recognize a small number of T. cruzi epitopes, preventing efficient pathogen elimination (Rodrigues et al., 2009). Immunodominant antigens can be selected by several factors, including stable MHC-peptide complexes on antigen-presenting cells, the affinity of T-cell receptors, as well as the abundance of parasite epitopes (Yewdell and Bennink, 1999;Goto et al., 2008;Rodrigues et al., 2009). Parasite trans-sialidases are among the significant known immunodominant targets for CD8 + T cells (Table 2), (Martin et al., 2006). ...
Chapter
Trypanosoma cruzi is a vector-borne protozoan parasite that causes Chagas disease (CD) (also named American trypanosomiasis), a chronic condition with notably diverse clinical outcomes. T. cruzi infection affects around 7 million people worldwide, reinforcing CD as a major global public health problem that is not only restricted to endemic countries. This chapter aims to unfold the complexity of Trypanosoma cruzi and its intricate interactions with vectors and hosts.
... Additionally, TRs have been observed in TS, mucins and MASP, and in several other hypothetical proteins exhibiting signal sequences or trans-membrane domains, some of which were tested as antigenic (78). Antigenicity of these tandems, and their wide distribution along CDS suggest that they constitute a general mechanism for including unspecific polyclonal immune responses. ...
Preprint
Full-text available
Although the genome of Trypanosoma cruzi , the causative agent of Chagas disease, was first made available in 2005, with additional strains reported later, the intrinsic genome complexity of this parasite (abundance of repetitive sequences and genes organized in tandem) has traditionally hindered high-quality genome assembly and annotation. This also limits diverse types of analyses that require high degree of precision. Long reads generated by third-generation sequencing technologies are particularly suitable to address the challenges associated with T. cruzi´s genome since they permit directly determining the full sequence of large clusters of repetitive sequences without collapsing them. This, in turn, allows not only accurate estimation of gene copy numbers but also circumvents assembly fragmentation. Here, we present the analysis of the genome sequences of two T. cruzi clones: the hybrid TCC (DTU TcVI) and the non-hybrid Dm28c (DTU TcI), determined by PacBio SMRT technology. The improved assemblies herein obtained permitted us to accurately estimate gene copy numbers, abundance and distribution of repetitive sequences (including satellites and retroelements). We found that the genome of T. cruzi is composed of a "core compartment" and a "disruptive compartment" which exhibit opposite gene and GC content composition. New tandem and disperse repetitive sequences were identified, including some located inside coding sequences. Additionally, homologous chromosomes were separately assembled, allowing us to retrieve haplotypes as separate contigs instead of a unique mosaic sequence. Finally, manual annotation of surface multigene families MUC and trans-sialidases allows now a better overview of these complex groups of genes.
... The literature shows that the best antigens are composed of repetitive motifs, which are present in various organisms and are characterized by the presence of two or more copies of amino acid sequences. This is because these sequences stimulate B cells by binding to these repetitive antigens, independent of T lymphocyte stimulation (Rosati et al., 2003;Goto et al., 2008Goto et al., , 2010; Valiente-Gabioud et al., 2011). Thus, proteins that exhibit these characteristics are strong candidates for use as diagnostic and vaccine targets. ...
Article
Full-text available
Visceral leishmaniosis is one of the most important zoonotic diseases on the planet and dogs are the main reservoir of canine visceral leishmaniosis (CVL) in endemic areas. They play an important role in human infection because in dogs the disease appears long time after infection, and they can move uncontrollably, contributing to disperse the parasite. To take the decision to treat the animals or for euthanasia, in an elimination programme, in order to reduce the parasitic load, it is necessary to diagnose correctly, having more effective tools. Our group has developed a new recombinant antigen-based kinesin-related gene of Leishmania braziliensis (Lbk39), which shows 59% amino acid identity to the L. infantum homologue. The Lbk39 gene was synthesized, inserted into the pLEXSY-sat2 vector and transfected into L. tarentolae cells by electroporation. The recombinant protein was secreted in the culture with a C-terminal histidine marker, purified, generating a product at 337.68 μ g mL ⁻¹ . A total of 152 sera from dog's endemic and non-endemic areas were used, being 78 positives and 75 negatives. The antigen Lbk39 showed 100% sensitivity and 96.1% specificity. We compared this antigen with other antigens such as total extract of the parasite, TRDPP, and our data indicate that Lbk39 has potential application in the diagnosis of CVL through antibody detection.
Article
Full-text available
Large scale mapping of antigens and epitopes is pivotal for developing immunotherapies but challenging, especially for eukaryotic pathogens, due to their large genomes. Here, we developed an integrated platform for genome phage display (gPhage) to show that unbiased libraries of the eukaryotic parasite Trypanosoma cruzi enable the identification of thousands of antigens recognized by serum samples from Chagas disease patients. Because most of these antigens are hypothetical proteins, gPhage provides evidence of their expression during infection. We built and validated a comprehensive map of Chagas disease antibody response to show how linear and putative conformation epitopes, many rich in repetitive elements, allow the parasite to evade a buildup of neutralizing antibodies directed against protein domains that mediate infection pathogenesis. Thus, the gPhage platform is a reproducible and effective tool for rapid simultaneous identification of epitopes and antigens, not only in Chagas disease, but perhaps also in globally emerging/reemerging acute pathogens.
Article
Full-text available
The DNA sequence of the gene for type 6 M protein of Streptococcus pyogenes contains two extended tandem repeat regions and one nontandem repeat region. We suggest that the duplication and deletion of these repeats generates the observed diversity in size and sequence among the family of M proteins in the group A streptococci. In addition, the DNA sequence reveals the presence of a 42-amino-acid signal peptide, a region rich in proline that is thought to be located in the cell wall, and a membrane anchor sequence at the carboxyl-terminal end of the protein. Signals similar to the consensus sequences recognized for the initiation of transcription and translation in Gram-positive bacteria have been identified in the DNA sequence. Codon usage is similar to that of other Gram-positive bacteria and significantly different from that of Escherichia coli.
Article
Full-text available
Whole-genome sequencing of the protozoan pathogen Trypanosoma cruzi revealed that the diploid genome contains a predicted 22,570 proteins encoded by genes, of which 12,570 represent allelic pairs. Over 50% of the genome consists of repeated sequences, such as retrotransposons and genes for large, families of surface molecules, which include trans-sialidases, mucins, gp63s, and a large novel family (>1300 copies) of mucin-associated surface protein (MASP) genes. Analyses of the T. cruzi, T. brucei, and Leishmania major (Tritryp) genomes imply differences from other eukaryotes in DNA repair and initiation of replication and reflect their unusual mitochondrial DNA. Although the Tritryp lack several classes of signaling molecules, their kinomes contain a large and diverse set of protein kinases and phosphatases; their size and diversity imply previously unknown interactions and regulatory processes, which may be targets for intervention.
Article
Full-text available
A 2.91-billion base pair (bp) consensus sequence of the euchromatic portion of the human genome was generated by the whole-genome shotgun sequencing method. The 14.8-billion bp DNA sequence was generated over 9 months from 27,271,853 high-quality sequence reads (5.11-fold coverage of the genome) from both ends of plasmid clones made from the DNA of five individuals. Two assembly strategies—a whole-genome assembly and a regional chromosome assembly—were used, each combining sequence data from Celera and the publicly funded genome effort. The public data were shredded into 550-bp segments to create a 2.9-fold coverage of those genome regions that had been sequenced, without including biases inherent in the cloning and assembly procedure used by the publicly funded group. This brought the effective coverage in the assemblies to eightfold, reducing the number and size of gaps in the final assembly over what would be obtained with 5.11-fold coverage. The two assembly strategies yielded very similar results that largely agree with independent mapping data. The assemblies effectively cover the euchromatic regions of the human chromosomes. More than 90% of the genome is in scaffold assemblies of 100,000 bp or more, and 25% of the genome is in scaffolds of 10 million bp or larger. Analysis of the genome sequence revealed 26,588 protein-encoding transcripts for which there was strong corroborating evidence and an additional ∼12,000 computationally derived genes with mouse matches or other weak supporting evidence. Although gene-dense clusters are obvious, almost half the genes are dispersed in low G+C sequence separated by large tracts of apparently noncoding sequence. Only 1.1% of the genome is spanned by exons, whereas 24% is in introns, with 75% of the genome being intergenic DNA. Duplications of segmental blocks, ranging in size up to chromosomal lengths, are abundant throughout the genome and reveal a complex evolutionary history. Comparative genomic analysis indicates vertebrate expansions of genes associated with neuronal function, with tissue-specific developmental regulation, and with the hemostasis and immune systems. DNA sequence comparisons between the consensus sequence and publicly funded genome data provided locations of 2.1 million single-nucleotide polymorphisms (SNPs). A random pair of human haploid genomes differed at a rate of 1 bp per 1250 on average, but there was marked heterogeneity in the level of polymorphism across the genome. Less than 1% of all SNPs resulted in variation in proteins, but the task of determining which SNPs have functional consequences remains an open challenge.
Article
Full-text available
Leishmania species cause a spectrum of human diseases in tropical and subtropical regions of the world. We have sequenced the 36 chromosomes of the 32.8-megabase haploid genome of Leishmania major (Friedlin strain) and predict 911 RNA genes, 39 pseudogenes, and 8272 protein-coding genes, of which 36% can be ascribed a putative function. These include genes involved in host-pathogen interactions, such as proteolytic enzymes, and extensive machinery for synthesis of complex surface glycoconjugates. The organization of protein-coding genes into long, strand-specific, polycistronic clusters and lack of general transcription factors in the L. major, Trypanosoma brucei, and Trypanosoma cruzi (Tritryp) genomes suggest that the mechanisms regulating RNA polymerase II–directed transcription are distinct from those operating in other eukaryotes, although the trypanosomatids appear capable of chromatin remodeling. Abundant RNA-binding proteins are encoded in the Tritryp genomes, consistent with active posttranscriptional regulation of gene expression.
Article
Full-text available
African trypanosomes cause human sleeping sickness and livestock trypanosomiasis in sub-Saharan Africa. We present the sequence and analysis of the 11 megabase-sized chromosomes of Trypanosoma brucei . The 26-megabase genome contains 9068 predicted genes, including ~900 pseudogenes and ~1700 T. brucei –specific genes. Large subtelomeric arrays contain an archive of 806 variant surface glycoprotein (VSG) genes used by the parasite to evade the mammalian immune system. Most VSG genes are pseudogenes, which may be used to generate expressed mosaic genes by ectopic recombination. Comparisons of the cytoskeleton and endocytic trafficking systems with those of humans and other eukaryotic organisms reveal major differences. A comparison of metabolic pathways encoded by the genomes of T. brucei , T. cruzi , and Leishmania major reveals the least overall metabolic capability in T. brucei and the greatest in L. major . Horizontal transfer of genes of bacterial origin has contributed to some of the metabolic differences in these parasites, and a number of novel potential drug targets have been identified.
Article
Full-text available
In this study, the occurrence of repeated DNA sequences in the chromosome of Mycobacterium tuberculosis was investigated systematically. By screening a M. tuberculosis lambda gt-11 gene library with labeled total chromosomal DNA, five strongly hybridizing recombinants were selected, and these contained DNA sequences that were present in multiple copies in the chromosome of M. tuberculosis. These recombinants all contained repeated sequences belonging to a single family of repetitive DNA, which shares homology with a previously described repeated sequence present in recombinant pPH7301. Sequences analysis of pPH7301 showed the presence of a 10-bp sequence that was tandemly repeated and invariably separated by 5-bp unique spacer sequences. Southern blot analysis revealed that the majority of the repeated DNA in M. tuberculosis is composed of this family of repetitive DNA. Because the 10-bp repeats are slightly heterogeneous in sequence, we designated this DNA as a major polymorphic tandem repeat, MPTR. The presence of this repeated sequence in various other mycobacterial species was investigated. Among the MPTR-containing mycobacterial species the chromosomal location of the repetitive DNA is highly variable. The potential use of this polymorphism in the epidemiology of mycobacterioses is discussed.
Article
Full-text available
A gene sequence encoding an immunodominant protein with a repetitive epitope from the protozoan Trypanosoma cruzi, the causative agent of Chagas disease, was cloned and expressed. The identified 10-amino acid repeat is present within a high-molecular-weight trypomastigote antigen that appears specific to and conserved among T. cruzi isolates. More importantly, greater than 95% of T. cruzi infection sera, including both chronic and acute Chagas disease, contained elevated levels of antibody to a 15-amino acid synthetic peptide bearing the repetitive B-cell epitope. Considering the wide diversity of T. cruzi parasites, as well as the broad spectrum of clinical manifestations of Chagas disease, such a prevalent immune response among patients is significant and applicable to the control of Chagas disease through the diagnosis of T. cruzi infection.
Article
Full-text available
Anaplasmosis is one of several tick-borne diseases severely constraining cattle production and usage in many parts of the world. Cattle can be protected from anaplasmosis by immunization with major surface protein 1, a surface protein of Anaplasma marginale carrying a neutralization-sensitive epitope. Marked size polymorphisms exist among different isolates of A. marginale in the AmF105 subunit of major surface protein 1, yet all isolates still contain the neutralization-sensitive epitope. To clarify the basis for these observations, the mspl alpha gene encoding AmF105 was cloned from four isolates and sequenced. The encoded polypeptides share a high degree of overall homology between isolates but contain a domain with various numbers of tandemly repeated sequences and three regions of clustered amino acid substitutions outside the repeat domain. The polypeptide size differences are completely explained by the variations in the numbers of tandem repeat units. We have mapped the neutralization-sensitive epitope to a sequence that is present within each repeat unit. These results identify a basis for size polymorphisms of the surface polypeptide antigen concomitant with B-cell epitope conservation in rickettsiae.
Article
All the protein sequences from SWISS-PROT database were analyzed for occurrence of single amino acid repeats, tandem oligo-peptide repeats, and periodically conserved amino acids. Single amino acid repeats of glutamine, serine, glutamic acid, glycine, and alanine seem to be tolerated to a considerable extent in many proteins. Tandem oligo-peptide repeats of different types with varying levels of conservation were detected in several proteins and found to be conspicuous, particularly in structural and cell surface proteins. It appears that repeated sequence patterns may be a mechanism that provides regular arrays of spatial and functional groups, useful for structural packing or for one to one interactions with target molecules. To facilitate further explorations, a database of Tandem Repeats in Protein Sequences (TRIPS) has been developed and is available at URL: http://www.ncl-india.org/trips.