Skip to main content
Erschienen in: Malaria Journal 1/2015

Open Access 01.12.2015 | Research

Diversity of malaria parasites in great apes in Gabon

verfasst von: Larson Boundenga, Benjamin Ollomo, Virginie Rougeron, Lauriane Yacka Mouele, Bertrand Mve-Ondo, Lucrèce M Delicat-Loembet, Nancy Diamella Moukodoum, Alain Prince Okouga, Céline Arnathau, Eric Elguero, Patrick Durand, Florian Liégeois, Vanina Boué, Peggy Motsch, Guillaume Le Flohic, Alphonse Ndoungouet, Christophe Paupy, Cheikh Tidiane Ba, Francois Renaud, Franck Prugnolle

Erschienen in: Malaria Journal | Ausgabe 1/2015

Abstract

Background

Until 2009, the Laverania subgenus counted only two representatives: Plasmodium falciparum and Plasmodium reichenowi. The recent development of non-invasive methods allowed re-exploration of plasmodial diversity in African apes. Although a large number of great ape populations have now been studied regarding Plasmodium infections in Africa, there are still vast areas of their distribution that remained unexplored. Gabon constitutes an important part of the range of western central African great ape subspecies (Pan troglodytes troglodytes and Gorilla gorilla gorilla), but has not been studied so far. In the present study, the diversity of Plasmodium species circulating in great apes in Gabon was analysed.

Methods

The analysis of 1,261 faecal samples from 791 chimpanzees and 470 gorillas collected from 24 sites all over Gabon was performed. Plasmodium infections were characterized by amplification and sequencing of a portion of the Plasmodium cytochrome b gene.

Results

The analysis of the 1,261 samples revealed that at least six Plasmodium species circulate in great apes in Gabon (Plasmodium praefalciparum, Plasmodium gorA (syn Plasmodium adleri), Plasmodium gorB (syn Plasmodium blacklocki) in gorillas and Plasmodium gaboni, P. reichenowi and Plasmodium billcollinsi in chimpanzees). No new phylogenetic lineages were discovered. The average infection rate was 21.3% for gorillas and 15.4% for chimpanzees. A logistic regression showed that the probability of infection was significantly dependent on the freshness of the droppings but not of the host species or of the average pluviometry of the months of collection.
Hinweise
Benjamin Ollomo and Franck Prugnolle contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

LB, BO, FP, VB, VR, APO, FL, AN, GL, PM, and CP contributed to the acquisition of samples in fieldwork; LB, BO, FP, APO, NDM, BMO, LMDL, LYM, CA, EE, PD, CTB, PM, GL, CP, and FR analysed and interpreted the data; LB, BO, CTB, FR, and FP conducted and supervised this work; LB, BO, FP, and FR wrote this paper. All authors read and approved the final manuscript.

Background

Plasmodium falciparum is a protozoan parasite responsible for malaria in humans. Among the five parasites infecting humans (Plasmodium falciparum, Plasmodium malariae, Plasmodium ovale, Plasmodium vivax and Plasmodium knowlesi), P. falciparum is by far the most virulent, responsible every year for approximately 207 million clinical cases and 627,000 deaths in the world [1], of which 98% are in sub-Saharan Africa [2-5]. Malaria is proving to be an obstacle that can slow down economic prosperity in many tropical countries, particularly in Africa [3].
Plasmodium falciparum belongs to the subgenus Laverania, which up to 2009 included only two known representatives: P. falciparum and Plasmodium reichenowi, a parasite from chimpanzees. Since 2009, thanks to the use of molecular tools for species identification and the development of non-invasive methods, several studies re-explored the diversity of Plasmodium species circulating in non-human primates in Africa, especially great apes (gorillas and chimpanzees) [6-8]. These studies revealed the existence of several lineages/species related to P. falciparum, deeply modifying the comprehension of the evolution of this parasite and of Laverania more generally. Four Laverania species are now recognized to infect chimpanzees: P. reichenowi, Plasmodium billcollinsi, Plasmodium gaboni and Plasmodium billbrayi [9-11]. For gorillas, there are three species: Plasmodium praefalciparum (the closest relative of P. falciparum), Plasmodium gorB (syn-Plasmodium blacklocki) and Plasmodium gorA (syn-Plasmodium adleri) [2,12]. Great apes have also been shown to be infected with species of the subgenus Plasmodium (non-Laverania): P. malariae-like, P. ovale-like and P. vivax-like parasites [8,13,14].
Was the entire diversity of Plasmodium species circulating in great apes in Africa discovered? Although a large number of great ape populations in Africa have now been studied regarding Plasmodium infections, there are still vast areas of their geographic distribution that remain unexplored. This is the case, for instance, for the western, central African populations of chimpanzees and gorillas (Pan troglodytes troglodytes and Gorilla gorilla gorilla). Although the range of both species covers all Gabon, half the surface of the Republic of the Congo, the south of Cameroon (south of the Sanaga river) and south of the Central African Republic, Plasmodium infections were almost only studied in populations from Cameroon, making about two-thirds of their range still unexplored [2,3,12].
In this study, using the second largest bank of faecal samples studied so far (more than 1,200 faecal samples), the diversity of Plasmodium species circulating in the great ape populations of Gabon was analysed. An investigation of the ecological factors susceptible to influence the detection of Plasmodium from these non-invasive samples was also performed.

Methods

Origin of faecal samples

Faecal samples of chimpanzees and gorillas were collected in 24 sites in Gabon from 2010 to 2014 (Figure 1a and Table 1). In the field, the origin of the faeces (chimpanzee or gorilla) was deduced according to cues such as the type of nest near which they were found, footprints, texture, and odours. Freshness of the faeces (>24or <24 hours post excretion) was also estimated based on the freshness of surrounding nests (when present), texture, colours, humidity, and level of degradation. All samples were preserved in RNAlater® (Life technologies, USA) and conserved at the CIRMF at -80°C. Their origin (chimpanzee or gorilla) was confirmed by mitochondrial DNA analysis as previously described [15,16]. This investigation was approved by the Government of the Republic of Gabon and with the authorization of the Agence Nationale des Parcs Nationaux (ANPN). In total, 791 faecal samples of chimpanzees and 470 of gorillas were collected and analysed.
Table 1
List of collection sites, abbreviations and geographical coordinates in Gabon
Sites
Abbreviations
Coordinates
(Degree, minute, second)
Lope
LP
S0°13′21.5″
E11°36′ 37.5″
Lope-Mikongo
LPM
S0°18′27.2″
E11°40′10.3″
Tsouba
TB
S1°09′54.2″
E14°26′46.8″
Makande
MD
S0°40′53.7″
E11°55′34.4″
Langoue
LG
N0°00′05.8″
E12°27′25.9″
Parc Ivindo-Langoue
PIL
S0°11′23.0″
E12°34′58.4″
Parc Ivindo-Iret-Kongou-Djidji
PIKD
N0°30′05.2″
E12°48′ 03.4″
Makatamangoye 1
MAK1
S0°08′39.3″
E13°36′47.6″
Monts De Cristal
MC
N0°40′15.4″
E10°24′54.2″
Parc Ivindo
PI
N0°23′24.8″
E12°41′33.1″
Djidji
DJ
N0°10′41.2″
E12°43′51.8″
Mwagna
MG
N0°38′53.5″
E13°52′08.2″
Boumango
BG
S1°43′36.0″
E14°03′10.0″
Malouma
ML
N0°39′01.6″
E13°52′17.2″
Lope 3
LP3
S0°19′32.4″
E11°37′23.6″
Gabonville
GV
N1°46′55.7″
E11°56′58.4″
Tomassi
TM
N1° 06′37.0″
E11°42′42.4″
Iyokomilieu
IYO
N0°02′54.1″
E13°36′05.6″
Boue
BE
S0°11′52. 7″
E12°02′01.8″
Parc de Loango
PL
S1°59′54.8″
E9°27′10.5″
Waka
WK
S1°07′57.3″
E11°08′30.8″
Bakoumba
BKB
S1°45′47.8″
E12°57′06.2″
Lope-Nord
LPN
N0°18′52.1″
E12°34′37.7″
Konossaville
KSV
N1°40′23.9″
E12°04′ 09.7″

Extraction of DNA and PCR

Faecal DNA was extracted using the QIAamp DNA Stool Mini Kit (Qiagen, Courteboeuf, France) as previously described [17] and Plasmodium infections were determined after amplification of a portion of Plasmodium mitochondrial genome (cytochrome b: cyt-b) as described in Prugnolle et al. [2]. All amplified products (10 μl) were run on 1.5% agarose gels in TAE buffer. The PCR-amplified products (956 bp) were used as templates for sequencing. DNA sequencing was performed by Eurofin MWG [18].

Species identification in mixed infections

When sequence chromatograms showed multiple peaks (heterozygous base calling), the program Mixed Sequences Reader (MSR) was used to determine if the isolates were mixed infected and by which species [19]. This program can directly analyse heterozygous base-calling fluorescence chromatograms and identify species in presence from a list of reference sequences (Table 2).
Table 2
Percentage of mixed infections detected from sequence chromatograms with multiple peaks using the program MSR (Mixed Sequences Reader)
Host
Percentage (%) of mixed infections
Associated species (n)
  
P. reichenowi + P. gaboni(6)
P. billcollinsi + P. gaboni(1)
Chimpanzees
20% (8/40)
P. reichenowi + P. billcollinsi(1)
Gorillas
28% (8/29)
P. adleri + P. praefalciparum(7)
P. adleri + P. blacklocky(1)
n: Number of mixed infection found.

Phylogenetic analyses

Phylogenetic analyses were performed using only cyt-b sequences derived from chromatograms with no ambiguous base calls. To examine the relationship of the cyt-b sequences obtained with the different Plasmodium species known so far, a phylogenetic tree was constructed using a set of reference sequences belonging to different Plasmodium species. Hosts and GenBank accession numbers for these reference sequences are given in Table 3. The multiple alignment of all partial cyt-b sequences (686 nucleotides) was done using ClustalW (v 1.8.1 in BioEdit v.7.0.9.0. software) [20]. Maximum likelihood (ML) tree construction was based on the cyt-b sequences. The best-fitting ML model under the Akaike Information Criterion was GTR (general time reversible) + ModelTest. [21] The highest-likelihood DNA tree and corresponding bootstrap support values were obtained by PhyML (freely available at the ATGC bioinformatics platform [22,23]) using NNI (nearest neighbour interchange) + SPR (sub-tree pruning regrafting) branch swapping and 100 bootstrap replicates [24].
Table 3
Accession numbers of the sequences of reference used in the phylogenetic tree
Accession number
Isolates
Species
Host species
References
HM235178
C1 BBptt238
P. reichenowi
Chimpanzee
Liu et al. [12]
HM235317
C2 LBptt176
P. gaboni
Chimpanzee
Liu et al. [12]
HM234979
C2 BBptt93
P. gaboni
Chimpanzee
Liu et al. [12]
HM234980
C1 BBptt93
P. reichenowi
Chimpanzee
Liu et al. [12]
HM234976
C3 BApts1413
P. billcollinsi
Chimpanzee
Liu et al. [12]
KC203544
EC4014_SGA500.11
P. falciparum
Human
Sundararaman et al. [25]
FJ895308
Isolate B
P. gaboni
Chimpanzee
Ollomo et al. [11]
GU045315
BQ642
P. reichenowi
Chimpanzee
Prugnolle et al. [2]
GQ355486
DRCJ
P. malariae
Bonobo
Krief et al. [26]
FJ409564
CPZcam91
P. ovale
Chimpanzee
Duval et al. [27]
KF591814
MRL49_FD_SGA1k.
P. vivax
Human
Liu et al. [14]
JQ345521
KN013
P. knowlesi
Human
Neoh Wan Fen et al. [28]
GU045317
BQ668
P. blacklocki
Gorilla
Prugnolle et al. [2]
GU045322
BQ638
P. adleri
Gorilla
Prugnolle et al. [2]
HM235386
G1 DDgor27
P. praefalciparum
Gorilla
Liu et al. [12]
HM235295
G3 DSgor24
P. blacklocki
Gorilla
Liu et al. [12]
HM235203
G1 DSgor86
P. praefalciparum
Gorilla
Liu et al. [12]
HM235059
G2 KKgor2638
P. adleri
Gorilla
Liu et al. [12]
JF923762
MO454
P. praefalciparum
C. nictitans
Prugnolle et al. [2]
GU815512
Louise
P. billcollinsi
Chimpanzee
Kaiser et al. [8]
GQ355478
UGF
P. billcollinsi
Chimpanzee
Krief et al. [26]
GQ355477
UGD
P. billcollinsi
Chimpanzee
Krief et al. [26]
AJ251941
-
P. reichenowi
Chimpanzee
Conway et al. [29]
JX893151
Clone39C
P. gaboni
Chimpanzee
Pacheco et al. [30]
JX893154
Clone20A
P. reichnowi
Chimpanzee
Pacheco et al. [30]

Statistical analyses

All statistical analyses were performed using R [31]. A logistic regression was used to analyse the variations among individuals in the infection status. In these models, the variable to be predicted was the presence/absence of a Plasmodium infection. The predictive variables were: (i) the site of collection (random effect); (ii) freshness of the faeces; (iii) host species; and, (iv) average pluviometry during months of collection (fixed effects). For the second predictive variable, faeces were subdivided into two groups: the faecal samples deposited less than 24 hours before collection and those collected after 24 hours. The host species corresponded to gorilla and chimpanzee. Finally, for each month of collection, the average Gabonese pluviometry (estimated from data collected from1960 to 1990) was retrieved from [32], which data were produced by the Climatic Research Unit (CRU) of University of East Anglia (UEA). Pluviometry was considered as a possible predictive variable because it is known to influence levels of infection in human foci [33,34].

GeneBank accession numbers published in this study

The sequences reported in this study were deposited in GenBank under the following accession numbers KP875428 to KP875480

Results

Plasmodium species infecting great apes in Gabon

Some 1,261 faecal samples from wild chimpanzees (n = 791) and gorillas (n = 470) from 24 sites were analysed (Figure 1a). Among them, 122 samples of chimpanzees (15.42%) and 100 of gorillas (21.28%) were detected positive to a Plasmodium infection by Cyt-b PCR. Sequences of quality (of sufficient size (>600 bp) and with a clear chromatogram) were obtained for 31% (n = 69) of the Cyt-b amplicons. Among them, sixteen showed multiple peaks and were identify as clear mixed infections by the program MSR. The frequency of mixed infections observed in chimpanzees and gorillas as estimated by the analysis of the chromatograms is given in Table 2. Phylogenetic analyses (Figure 2) revealed the presence of three Plasmodium species in chimpanzees (P. gaboni, P. reichenowi and P. billcollinsi) and three in gorillas (P. praefalciparum, P. gorA and P. gorB). Neither species of the subgenus Plasmodium (P. vivax-like, P. malariae-like and P. ovale-like) nor new phylogenetic lineages were found in these samples. Relative frequencies of each Plasmodium species in each site among positives are given in Figure 1a-c.
Over the entire dataset, logistic regressions revealed that the probability of infection was only significantly dependent on the variable ‘freshness of the stool’. Pluviometry as well as host species did not significantly explain the probability of infection (Table 4). As shown in Table 4, the probability of infection was higher in stools collected less than 24 hours after dropping than in the older ones. Overall, freshness of the stools did not significantly differ between chimpanzees and gorillas (p-value = 0.07).
Table 4
Results of the logistic regression
Variable
P-value
Odds ratio [CI 95% ]
Host species
0.051390
0.67 [0.503 to 0. 905]
Freshness of the faeces
0.006684
2.038[1.458 to 2. 849]
Pluviometry
0.581011
0.576 [0.429 to 0. 775]
The presence or absence of infection by Plasmodium was the variable to be predicted. Predictive variables were: host species, freshness of faeces (<24 or >24 hr) and pluviometry. CI95%: 95% Confidence Interval.

Discussion

In the last few years, several new Plasmodium species were discovered in African non-human primates, especially great apes [2,3,6,10,26]. These discoveries were made possible by the development of a non-invasive method allowing detection of Plasmodium infections from faecal samples [2,6], despite inherent problems of DNA degradation with this type of biological material. This issue was overcome by the use of mitochondrial sequences to amplify the parasite, which presents several advantages: 1) Mitochondrial DNA is in multiple copy inside parasites (unlike nuclear DNA) and 2) if properly chosen, small portions of the mitochondrial genome (as small as 200 bp), can contain enough phylogenetic information to identify the different Plasmodium species. This method is now one of the main methods used to analyse Plasmodium from wild non-human primates [2,7,12,25].
In the present study, analyses were performed on a set of 1,261 faecal samples collected all over Gabon from chimpanzees and gorillas. All Plasmodium species found belonged to the subgenus Laverania and were all previously identified in Pan troglodytes troglodytes and Gorilla gorilla gorilla, respectively [2,12]. No new phylogenetic lineage or species were identified. Surprisingly, no species of the subgenus Plasmodium (non-Laverania) were identified either. This is at odds with recent observations made from ape blood samples or infected sylvatic anopheline mosquitoes collected in Gabon showing the circulation of P. vivax-like parasites in the area. [13] These results are nevertheless congruent with those from Liu et al. [12]. Although they analysed 3,000 ape faecal samples from west and central Africa, they only obtained seven sequences of Plasmodium belonging to the subgenus Plasmodium (non-Laverania).
One main factor could explain why parasites of the subgenus Plasmodium were not detected and this is most likely linked to the nature of the primers used to perform PCRs. Indeed, as in the study of Liu et al. [12], the primers used were specifically designed to amplify sequences of Laverania parasites. As a consequence, several nucleotides of differences separated them from the homologous sequences in P. vivax, P. malariae and P. ovale, thus very likely reducing the sensitivity of this PCR to detect non-Laverania species. In addition, such problem might have been amplified by the presence of co-infections with Laverania species. Indeed, it has been demonstrated that in case of co-infection, the PCR tends to favour the amplification of the parasite with the best matching sequence to the primers [35,36]. Recently, Liu et al. [14] solved this problem by designing primers specific to P. vivax. Out of the 3,000 samples previously analysed and re-analysed with other samples, they finally detected more than 87 P. vivax infections.
In this study, no Laverania species were found to infect both hosts (gorillas and chimpanzees). This reinforces the hypothesis that Laverania lineages infect specific hosts [10], a specificity that could be associated to specific ligand/receptor interactions occurring in the vertebrate host, as suggested by several studies [37], or by ecological factors such as the trophic preferences of the vectors [38] or the fact that gorillas’ and chimpanzees’ home ranges might not overlap in space and time. Additional studies would be needed to disentangle these different possibilities.
No human Plasmodium species were found. This result is congruent with other studies performed so far on wild populations of apes [8,12,39] thus confirming that, contrarily to what some authors have feared [7,40], great apes do not (and will certainly never) constitute reservoirs of Plasmodium, in particular P. falciparum, for humans. The fact that their populations are rapidly declining [41,42] is unfortunately another element in support of this prediction. Finally, no evidence of ape-to-human transfers of Laverania species was ever recorded despite efforts to find them [37]. The only documented record of this kind of transfer, in a natural context, was for a P. vivax-like (non-Laverania) parasite [13].
Regarding the prevalence of infections, more than 15% of the chimpanzee and 21% of the gorilla samples were positive to Plasmodium. Infections were detected in 17out of 23sites for chimpanzees and 16 out of 24 for gorillas. Global rates of infection found in this study are similar to those found by Prugnolle et al. [2], Kaiser et al. [8], and Liu et al. [12] in other areas. As previously discussed [2,12], it is very likely that the accurate rates of infection are higher, because the detection of Plasmodium in this kind of biological material (faecal) is expected to be less sensitive than in blood, as it is the case for urine and saliva [12,43,44], due to sample degradation or repeated sampling (faecal samples from the same individual may have been collected several times). The effect of sample degradation (and hence DNA degradation) is evident here when comparing the rates of infection detected in the faeces that were collected before and after 24 hours post excretion. The freshest (and so the less degraded) faecal samples significantly present more Plasmodium infections than the other ones (odds ratio = 2.038).

Acknowledgements

Authors thank the three reviewers for their constructive comments. The study was funded by Centre International de Recherches Médicales de Franceville (CIRMF, Gabon), Centre National de la Recherche Scientifique (CNRS, France), Institut de Recherche pour le Développement (IRD, France) and Agence Nationale de la Recherche (ANR, France, grant ORIGIN JCJC 012). We thank all the persons that were involved in the sampling as well as the Gabonese national agency of the national parks (authorization of collection: N° AE13009/PR/ANPN/SE/CS/AEPN). We also thank Bouanga Miriame for her help.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

LB, BO, FP, VB, VR, APO, FL, AN, GL, PM, and CP contributed to the acquisition of samples in fieldwork; LB, BO, FP, APO, NDM, BMO, LMDL, LYM, CA, EE, PD, CTB, PM, GL, CP, and FR analysed and interpreted the data; LB, BO, CTB, FR, and FP conducted and supervised this work; LB, BO, FP, and FR wrote this paper. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat WHO. World malaria report 2013. Geneva: World Health Organization; 2014. p. 178. WHO. World malaria report 2013. Geneva: World Health Organization; 2014. p. 178.
2.
Zurück zum Zitat Prugnolle F, Ollomo B, Durand P, Yalcindag E, Arnathau C, Elguero E, et al. African monkeys are infected by Plasmodium falciparum nonhuman primate-specific strains. Proc Natl Acad Sci U S A. 2011;108:11948–53.CrossRefPubMedCentralPubMed Prugnolle F, Ollomo B, Durand P, Yalcindag E, Arnathau C, Elguero E, et al. African monkeys are infected by Plasmodium falciparum nonhuman primate-specific strains. Proc Natl Acad Sci U S A. 2011;108:11948–53.CrossRefPubMedCentralPubMed
3.
Zurück zum Zitat Rich SM, Leendertz FH, Xu G, LeBreton M, Djoko CF, Aminake MN, et al. The origin of malignant malaria. Proc Natl Acad Sci U S A. 2009;106:14902–7.CrossRefPubMedCentralPubMed Rich SM, Leendertz FH, Xu G, LeBreton M, Djoko CF, Aminake MN, et al. The origin of malignant malaria. Proc Natl Acad Sci U S A. 2009;106:14902–7.CrossRefPubMedCentralPubMed
4.
Zurück zum Zitat Jeffares DC, Pain A, Berry A, Cox AV, Stalker J, Ingle CE, et al. Genome variation and evolution of the malaria parasite Plasmodium falciparum. Nat Genet. 2007;39:120–5.CrossRefPubMedCentralPubMed Jeffares DC, Pain A, Berry A, Cox AV, Stalker J, Ingle CE, et al. Genome variation and evolution of the malaria parasite Plasmodium falciparum. Nat Genet. 2007;39:120–5.CrossRefPubMedCentralPubMed
6.
Zurück zum Zitat Prugnolle F, Ayala F, Ollomo B, Arnathau C, Durand P, Renaud F. Plasmodium falciparum is not as lonely as previously considered. Virulence. 2011;2:71–6.CrossRefPubMed Prugnolle F, Ayala F, Ollomo B, Arnathau C, Durand P, Renaud F. Plasmodium falciparum is not as lonely as previously considered. Virulence. 2011;2:71–6.CrossRefPubMed
7.
Zurück zum Zitat Duval L, Fourment M, Nerrienet E, Rousset D, Sadeuh SA, Goodman SM, et al. African apes as reservoirs of Plasmodium falciparum and the origin and diversification of the Laverania subgenus. Proc Natl Acad Sci U S A. 2010;107:10561–6.CrossRefPubMedCentralPubMed Duval L, Fourment M, Nerrienet E, Rousset D, Sadeuh SA, Goodman SM, et al. African apes as reservoirs of Plasmodium falciparum and the origin and diversification of the Laverania subgenus. Proc Natl Acad Sci U S A. 2010;107:10561–6.CrossRefPubMedCentralPubMed
8.
Zurück zum Zitat Kaiser M, Lowa A, Ulrich M, Ellerbrok H, Goffe AS, Blasse A, et al. Wild chimpanzees infected with 5 Plasmodium species. Emerg Infect Dis. 2010;16:1956–9.CrossRefPubMedCentralPubMed Kaiser M, Lowa A, Ulrich M, Ellerbrok H, Goffe AS, Blasse A, et al. Wild chimpanzees infected with 5 Plasmodium species. Emerg Infect Dis. 2010;16:1956–9.CrossRefPubMedCentralPubMed
9.
Zurück zum Zitat Snounou G, Escalante A, Kasenene J, Renia L, Gruner AC, Krief S. [Malaria in hominids](in French). Bull Acad Natl Med. 2011;195:1945–54.PubMed Snounou G, Escalante A, Kasenene J, Renia L, Gruner AC, Krief S. [Malaria in hominids](in French). Bull Acad Natl Med. 2011;195:1945–54.PubMed
10.
Zurück zum Zitat Rayner JC, Liu W, Peeters M, Sharp PM, Hahn BH. A plethora of Plasmodium species in wild apes: a source of human infection? Trends Parasitol. 2011;27:222–9.CrossRefPubMedCentralPubMed Rayner JC, Liu W, Peeters M, Sharp PM, Hahn BH. A plethora of Plasmodium species in wild apes: a source of human infection? Trends Parasitol. 2011;27:222–9.CrossRefPubMedCentralPubMed
11.
12.
Zurück zum Zitat Liu W, Li Y, Learn GH, Rudicell RS, Robertson JD, Keele BF, et al. Origin of the human malaria parasite Plasmodium falciparum in gorillas. Nature. 2010;467:420–5.CrossRefPubMedCentralPubMed Liu W, Li Y, Learn GH, Rudicell RS, Robertson JD, Keele BF, et al. Origin of the human malaria parasite Plasmodium falciparum in gorillas. Nature. 2010;467:420–5.CrossRefPubMedCentralPubMed
13.
Zurück zum Zitat Prugnolle F, Rougeron V, Becquart P, Berry A, Makanga B, Rahola N, et al. Diversity, host switching and evolution of Plasmodium vivax infecting African great apes. Proc Natl Acad Sci U S A. 2013;110:8123–8.CrossRefPubMedCentralPubMed Prugnolle F, Rougeron V, Becquart P, Berry A, Makanga B, Rahola N, et al. Diversity, host switching and evolution of Plasmodium vivax infecting African great apes. Proc Natl Acad Sci U S A. 2013;110:8123–8.CrossRefPubMedCentralPubMed
14.
Zurück zum Zitat Liu W, Li Y, Shaw KS, Learn GH, Plenderleith LJ, Malenke JA, et al. African origin of the malaria parasite Plasmodium vivax. Nat Commun. 2014;5:3346.PubMedCentralPubMed Liu W, Li Y, Shaw KS, Learn GH, Plenderleith LJ, Malenke JA, et al. African origin of the malaria parasite Plasmodium vivax. Nat Commun. 2014;5:3346.PubMedCentralPubMed
15.
Zurück zum Zitat Santiago ML, Lukasik M, Kamenya S, Li Y, Bibollet-Ruche F, Bailes E, et al. Foci of endemic simian immunodeficiency virus infection in wild-living eastern chimpanzees (Pan troglodytes schweinfurthii). J Virol. 2003;77:7545–62.CrossRefPubMedCentralPubMed Santiago ML, Lukasik M, Kamenya S, Li Y, Bibollet-Ruche F, Bailes E, et al. Foci of endemic simian immunodeficiency virus infection in wild-living eastern chimpanzees (Pan troglodytes schweinfurthii). J Virol. 2003;77:7545–62.CrossRefPubMedCentralPubMed
16.
Zurück zum Zitat Santiago ML, Range F, Keele BF, Li Y, Bailes E, Bibollet-Ruche F, et al. Simian immunodeficiency virus infection in free-ranging sooty mangabeys (Cercocebus atys atys) from the Tai Forest, Cote d’Ivoire: implications for the origin of epidemic human immunodeficiency virus type 2. J Virol. 2005;79:12515–27.CrossRefPubMedCentralPubMed Santiago ML, Range F, Keele BF, Li Y, Bailes E, Bibollet-Ruche F, et al. Simian immunodeficiency virus infection in free-ranging sooty mangabeys (Cercocebus atys atys) from the Tai Forest, Cote d’Ivoire: implications for the origin of epidemic human immunodeficiency virus type 2. J Virol. 2005;79:12515–27.CrossRefPubMedCentralPubMed
17.
Zurück zum Zitat Keele BF, Van Heuverswyn F, Li Y, Bailes E, Takehisa J, Santiago ML, et al. Chimpanzee reservoirs of pandemic and nonpandemic HIV-1. Science. 2006;313:523–6.CrossRefPubMedCentralPubMed Keele BF, Van Heuverswyn F, Li Y, Bailes E, Takehisa J, Santiago ML, et al. Chimpanzee reservoirs of pandemic and nonpandemic HIV-1. Science. 2006;313:523–6.CrossRefPubMedCentralPubMed
19.
Zurück zum Zitat Chang CT, Tsai CN, Tang CY, Chen CH, Lian JH, Hu CY, et al. Mixed sequence reader: a program for analyzing DNA sequences with heterozygous base calling. Sci World J. 2012;2012:365104. Chang CT, Tsai CN, Tang CY, Chen CH, Lian JH, Hu CY, et al. Mixed sequence reader: a program for analyzing DNA sequences with heterozygous base calling. Sci World J. 2012;2012:365104.
20.
Zurück zum Zitat Hall T. BioEdit: A user-friendly biological sequence alignment editor and analysis program for Windows 95/98/NT. Nucleic Acids Symp Ser. 1999;41:45–98. Hall T. BioEdit: A user-friendly biological sequence alignment editor and analysis program for Windows 95/98/NT. Nucleic Acids Symp Ser. 1999;41:45–98.
21.
Zurück zum Zitat Posada D, Crandall KA. MODELTEST: testing the model of DNA substitution. Bioinformatics. 1998;14:817–8.CrossRefPubMed Posada D, Crandall KA. MODELTEST: testing the model of DNA substitution. Bioinformatics. 1998;14:817–8.CrossRefPubMed
22.
Zurück zum Zitat Dereeper A, Guignon V, Blanc G, Audic S, Buffet S, Chevenet F, et al. Phylogeny.fr: robust phylogenetic analysis for the non-specialist. Nucleic Acids Res. 2008;36:465–9.CrossRef Dereeper A, Guignon V, Blanc G, Audic S, Buffet S, Chevenet F, et al. Phylogeny.fr: robust phylogenetic analysis for the non-specialist. Nucleic Acids Res. 2008;36:465–9.CrossRef
23.
Zurück zum Zitat Guindon S, Gascuel O. A simple, fast, and accurate algorithm to estimate large phylogenies by maximum likelihood. Syst Biol. 2003;52:696–704.CrossRefPubMed Guindon S, Gascuel O. A simple, fast, and accurate algorithm to estimate large phylogenies by maximum likelihood. Syst Biol. 2003;52:696–704.CrossRefPubMed
24.
Zurück zum Zitat Guindon S, Dufayard JF, Lefort V, Anisimova M, Hordijk W, Gascuel O. New algorithms and methods to estimate maximum-likelihood phylogenies: assessing the performance of PhyML 3.0. Syst Biol. 2010;59:307–21.CrossRefPubMed Guindon S, Dufayard JF, Lefort V, Anisimova M, Hordijk W, Gascuel O. New algorithms and methods to estimate maximum-likelihood phylogenies: assessing the performance of PhyML 3.0. Syst Biol. 2010;59:307–21.CrossRefPubMed
25.
Zurück zum Zitat Sundararaman SA, Liu W, Keele BF, Learn GH, Bittinger K, Mouacha F, et al. Plasmodium falciparum-like parasites infecting wild apes in southern Cameroon do not represent a recurrent source of human malaria. Proc Natl Acad Sci U S A. 2013;110:7020–5.CrossRefPubMedCentralPubMed Sundararaman SA, Liu W, Keele BF, Learn GH, Bittinger K, Mouacha F, et al. Plasmodium falciparum-like parasites infecting wild apes in southern Cameroon do not represent a recurrent source of human malaria. Proc Natl Acad Sci U S A. 2013;110:7020–5.CrossRefPubMedCentralPubMed
26.
Zurück zum Zitat Krief S, Escalante AA, Pacheco MA, Mugisha L, Andre C, Halbwax M, et al. On the diversity of malaria parasites in African apes and the origin of Plasmodium falciparum from Bonobos. PLoS Pathog. 2010;6:e1000765.CrossRefPubMedCentralPubMed Krief S, Escalante AA, Pacheco MA, Mugisha L, Andre C, Halbwax M, et al. On the diversity of malaria parasites in African apes and the origin of Plasmodium falciparum from Bonobos. PLoS Pathog. 2010;6:e1000765.CrossRefPubMedCentralPubMed
27.
Zurück zum Zitat Duval L, Nerrienet E, Rousset D, Sadeuh Mba SA, Houze S, Fourment M, et al. Chimpanzee malaria parasites related to Plasmodium ovale in Africa. PLoS One. 2009;4:e5520.CrossRefPubMedCentralPubMed Duval L, Nerrienet E, Rousset D, Sadeuh Mba SA, Houze S, Fourment M, et al. Chimpanzee malaria parasites related to Plasmodium ovale in Africa. PLoS One. 2009;4:e5520.CrossRefPubMedCentralPubMed
28.
Zurück zum Zitat Joveen-Neoh WF, Chong KL, Wong CM, Lau TY. Incidence of malaria in the interior division of sabah, malaysian borneo, based on nested PCR. J Parasitol Res. 2011;2011:104284.CrossRefPubMedCentralPubMed Joveen-Neoh WF, Chong KL, Wong CM, Lau TY. Incidence of malaria in the interior division of sabah, malaysian borneo, based on nested PCR. J Parasitol Res. 2011;2011:104284.CrossRefPubMedCentralPubMed
29.
Zurück zum Zitat Conway DJ, Fanello C, Lloyd JM, Al-Joubori BM, Baloch AH, Somanath SD, et al. Origin of Plasmodium falciparum malaria is traced by mitochondrial DNA. Mol Biochem Parasitol. 2000;111:163–71.CrossRefPubMed Conway DJ, Fanello C, Lloyd JM, Al-Joubori BM, Baloch AH, Somanath SD, et al. Origin of Plasmodium falciparum malaria is traced by mitochondrial DNA. Mol Biochem Parasitol. 2000;111:163–71.CrossRefPubMed
30.
Zurück zum Zitat Pacheco MA, Cranfield M, Cameron K, Escalante AA. Malarial parasite diversity in chimpanzees: the value of comparative approaches to ascertain the evolution of Plasmodium falciparum antigens. Malar J. 2013;12:328.CrossRefPubMedCentralPubMed Pacheco MA, Cranfield M, Cameron K, Escalante AA. Malarial parasite diversity in chimpanzees: the value of comparative approaches to ascertain the evolution of Plasmodium falciparum antigens. Malar J. 2013;12:328.CrossRefPubMedCentralPubMed
31.
Zurück zum Zitat R Development Core Team. R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing; 2008. URL: http://www.R-project.org. ISBN 3-900051-07-0. R Development Core Team. R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing; 2008. URL: http://​www.​R-project.​org. ISBN 3-900051-07-0.
33.
Zurück zum Zitat Robert V, Dieng H, Lochouran L, Traore SF, Trape JF, Simondon F, et al. [Malaria transmission in the rural zone of Niakhar, Senegal](in French). Trop Med Int Health. 1998;3:667–77.CrossRefPubMed Robert V, Dieng H, Lochouran L, Traore SF, Trape JF, Simondon F, et al. [Malaria transmission in the rural zone of Niakhar, Senegal](in French). Trop Med Int Health. 1998;3:667–77.CrossRefPubMed
34.
Zurück zum Zitat Ouldabdallahi M, Ouldbezeid M, Diop C, Dem E, Lassana K. [Epidemiology of human schistosomiasis in Mauritania. The right bank of the Senegal River as model](in French). Bull Soc Pathol Exot. 2010;103:317–22.CrossRefPubMed Ouldabdallahi M, Ouldbezeid M, Diop C, Dem E, Lassana K. [Epidemiology of human schistosomiasis in Mauritania. The right bank of the Senegal River as model](in French). Bull Soc Pathol Exot. 2010;103:317–22.CrossRefPubMed
35.
Zurück zum Zitat Valkiunas G, Palinauskas V, Ilgunas M, Bukauskaite D, Dimitrov D, Bernotiene R, et al. Molecular characterization of five widespread avian haemosporidian parasites (Haemosporida), with perspectives on the PCR-based detection of haemosporidians in wildlife. Parasitol Res. 2014;113:2251–63.CrossRefPubMed Valkiunas G, Palinauskas V, Ilgunas M, Bukauskaite D, Dimitrov D, Bernotiene R, et al. Molecular characterization of five widespread avian haemosporidian parasites (Haemosporida), with perspectives on the PCR-based detection of haemosporidians in wildlife. Parasitol Res. 2014;113:2251–63.CrossRefPubMed
36.
Zurück zum Zitat Zehtindjiev P, Krizanauskiene A, Bensch S, Palinauskas V, Asghar M, Dimitrov D, et al. A new morphologically distinct avian malaria parasite that fails detection by established polymerase chain reaction-based protocols for amplification of the cytochrome B gene. J Parasitol. 2012;98:657–65.CrossRefPubMed Zehtindjiev P, Krizanauskiene A, Bensch S, Palinauskas V, Asghar M, Dimitrov D, et al. A new morphologically distinct avian malaria parasite that fails detection by established polymerase chain reaction-based protocols for amplification of the cytochrome B gene. J Parasitol. 2012;98:657–65.CrossRefPubMed
37.
Zurück zum Zitat Wanaguru M, Liu W, Hahn BH, Rayner JC, Wright GJ. RH5-Basigin interaction plays a major role in the host tropism of Plasmodium falciparum. Proc Natl Acad Sci U S A. 2013;110:20735–40.CrossRefPubMedCentralPubMed Wanaguru M, Liu W, Hahn BH, Rayner JC, Wright GJ. RH5-Basigin interaction plays a major role in the host tropism of Plasmodium falciparum. Proc Natl Acad Sci U S A. 2013;110:20735–40.CrossRefPubMedCentralPubMed
38.
Zurück zum Zitat Paupy C, Makanga B, Ollomo B, Rahola N, Durand P, Magnus J, et al. Anopheles moucheti and Anopheles vinckei are candidate vectors of ape Plasmodium parasites, including Plasmodium praefalciparum in Gabon. PLoS One. 2013;8:e57294.CrossRefPubMedCentralPubMed Paupy C, Makanga B, Ollomo B, Rahola N, Durand P, Magnus J, et al. Anopheles moucheti and Anopheles vinckei are candidate vectors of ape Plasmodium parasites, including Plasmodium praefalciparum in Gabon. PLoS One. 2013;8:e57294.CrossRefPubMedCentralPubMed
39.
Zurück zum Zitat Prugnolle F, Durand P, Neel C, Ollomo B, Ayala FJ, Arnathau C, et al. African great apes are natural hosts of multiple related malaria species, including Plasmodium falciparum. Proc Natl Acad Sci U S A. 2010;107:1458–63.CrossRefPubMedCentralPubMed Prugnolle F, Durand P, Neel C, Ollomo B, Ayala FJ, Arnathau C, et al. African great apes are natural hosts of multiple related malaria species, including Plasmodium falciparum. Proc Natl Acad Sci U S A. 2010;107:1458–63.CrossRefPubMedCentralPubMed
40.
Zurück zum Zitat Duval L, Ariey F. Ape Plasmodium parasites as a source of human outbreaks. Clin Microbiol Infect. 2012;18:528–32.CrossRefPubMed Duval L, Ariey F. Ape Plasmodium parasites as a source of human outbreaks. Clin Microbiol Infect. 2012;18:528–32.CrossRefPubMed
41.
Zurück zum Zitat Prado-Martinez J, Sudmant PH, Kidd JM, Li H, Kelley JL, Lorente-Galdos B, et al. Great ape genetic diversity and population history. Nature. 2013;499:471–5.CrossRefPubMed Prado-Martinez J, Sudmant PH, Kidd JM, Li H, Kelley JL, Lorente-Galdos B, et al. Great ape genetic diversity and population history. Nature. 2013;499:471–5.CrossRefPubMed
42.
Zurück zum Zitat Campbell G, Kuehl H, N’Goran Kouame P, Boesch C. Alarming decline of West African chimpanzees in Cote d’Ivoire. Curr Biol. 2008;18:R903–4.CrossRefPubMed Campbell G, Kuehl H, N’Goran Kouame P, Boesch C. Alarming decline of West African chimpanzees in Cote d’Ivoire. Curr Biol. 2008;18:R903–4.CrossRefPubMed
43.
Zurück zum Zitat Nwakanma DC, Gomez-Escobar N, Walther M, Crozier S, Dubovsky F, Malkin E, et al. Quantitative detection of Plasmodium falciparum DNA in saliva, blood, and urine. J Infect Dis. 2009;199:1567–74.CrossRefPubMed Nwakanma DC, Gomez-Escobar N, Walther M, Crozier S, Dubovsky F, Malkin E, et al. Quantitative detection of Plasmodium falciparum DNA in saliva, blood, and urine. J Infect Dis. 2009;199:1567–74.CrossRefPubMed
44.
Zurück zum Zitat Mharakurwa S, Simoloka C, Thuma PE, Shiff CJ, Sullivan DJ. PCR detection of Plasmodium falciparum in human urine and saliva samples. Malar J. 2006;5:103.CrossRefPubMedCentralPubMed Mharakurwa S, Simoloka C, Thuma PE, Shiff CJ, Sullivan DJ. PCR detection of Plasmodium falciparum in human urine and saliva samples. Malar J. 2006;5:103.CrossRefPubMedCentralPubMed
Metadaten
Titel
Diversity of malaria parasites in great apes in Gabon
verfasst von
Larson Boundenga
Benjamin Ollomo
Virginie Rougeron
Lauriane Yacka Mouele
Bertrand Mve-Ondo
Lucrèce M Delicat-Loembet
Nancy Diamella Moukodoum
Alain Prince Okouga
Céline Arnathau
Eric Elguero
Patrick Durand
Florian Liégeois
Vanina Boué
Peggy Motsch
Guillaume Le Flohic
Alphonse Ndoungouet
Christophe Paupy
Cheikh Tidiane Ba
Francois Renaud
Franck Prugnolle
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Malaria Journal / Ausgabe 1/2015
Elektronische ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-015-0622-6

Weitere Artikel der Ausgabe 1/2015

Malaria Journal 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.