Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.811
Filter
1.
Nature ; 631(8019): 125-133, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38867050

ABSTRACT

Malaria-causing protozoa of the genus Plasmodium have exerted one of the strongest selective pressures on the human genome, and resistance alleles provide biomolecular footprints that outline the historical reach of these species1. Nevertheless, debate persists over when and how malaria parasites emerged as human pathogens and spread around the globe1,2. To address these questions, we generated high-coverage ancient mitochondrial and nuclear genome-wide data from P. falciparum, P. vivax and P. malariae from 16 countries spanning around 5,500 years of human history. We identified P. vivax and P. falciparum across geographically disparate regions of Eurasia from as early as the fourth and first millennia BCE, respectively; for P. vivax, this evidence pre-dates textual references by several millennia3. Genomic analysis supports distinct disease histories for P. falciparum and P. vivax in the Americas: similarities between now-eliminated European and peri-contact South American strains indicate that European colonizers were the source of American P. vivax, whereas the trans-Atlantic slave trade probably introduced P. falciparum into the Americas. Our data underscore the role of cross-cultural contacts in the dissemination of malaria, laying the biomolecular foundation for future palaeo-epidemiological research into the impact of Plasmodium parasites on human history. Finally, our unexpected discovery of P. falciparum in the high-altitude Himalayas provides a rare case study in which individual mobility can be inferred from infection status, adding to our knowledge of cross-cultural connectivity in the region nearly three millennia ago.


Subject(s)
DNA, Ancient , Genome, Mitochondrial , Genome, Protozoan , Malaria , Plasmodium , Female , Humans , Male , Altitude , Americas/epidemiology , Asia/epidemiology , Biological Evolution , Disease Resistance/genetics , DNA, Ancient/analysis , Europe/epidemiology , Genome, Mitochondrial/genetics , Genome, Protozoan/genetics , History, Ancient , Malaria/parasitology , Malaria/history , Malaria/transmission , Malaria/epidemiology , Malaria, Falciparum/epidemiology , Malaria, Falciparum/history , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Malaria, Vivax/epidemiology , Malaria, Vivax/history , Malaria, Vivax/parasitology , Malaria, Vivax/transmission , Plasmodium/genetics , Plasmodium/classification , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Plasmodium malariae/genetics , Plasmodium malariae/isolation & purification , Plasmodium vivax/genetics , Plasmodium vivax/isolation & purification
2.
Parasitol Res ; 123(6): 252, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38922536

ABSTRACT

Avian haemosporidians of the genera Plasmodium and Haemoproteus are a group of widely distributed blood parasites that can negatively affect the fitness of their hosts. Colombia contains the greatest diversity of birds on the planet, but knowledge about the associations between haemosporidian and its avifauna is scarce and fragmented. We collected blood samples from 255 birds (203 residents and 52 neotropical migrants) belonging to 27 families and 108 species. The study was conducted in six localities in the inter-Andean valleys of the Cauca and Magdalena rivers. Parasites of the genera Plasmodium and Haemoproteus were identified in the samples by morphological and molecular analysis of a fragment of the mitochondrial gene cyt b. Among the samples, 9.3% (n = 24) were positive for Plasmodium or Haemoproteus. Co-infection with Plasmodium and Haemoproteus was found in Red-eyed Vireo. Seventeen haemosporidian lineages were identified, five of which were reported for the first time in resident birds (Common Ground Dove, Checker-throated Stipplethroat, Tropical Kingbird, Pale-breasted Thrush, and Ruddy-breasted Seedeater) and one in the Summer Tanager (neotropical migrant). The research results confirm the wide diversity of haemosporidian present in tropical lowlands and the possible role of neotropical migratory birds in dissemination on haemosporidian along their migratory routes.


Subject(s)
Bird Diseases , Birds , Haemosporida , Plasmodium , Protozoan Infections, Animal , Animals , Colombia/epidemiology , Haemosporida/classification , Haemosporida/isolation & purification , Haemosporida/genetics , Birds/parasitology , Bird Diseases/parasitology , Bird Diseases/epidemiology , Plasmodium/classification , Plasmodium/isolation & purification , Plasmodium/genetics , Protozoan Infections, Animal/parasitology , Protozoan Infections, Animal/epidemiology , Cytochromes b/genetics , Animal Migration , Phylogeny , Coinfection/parasitology , Coinfection/veterinary , Coinfection/epidemiology
3.
Open Biol ; 14(6): 230451, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38862023

ABSTRACT

Plasmodium species encode a unique set of six modular proteins named LCCL lectin domain adhesive-like proteins (LAPs) that operate as a complex and that are essential for malaria parasite transmission from mosquito to vertebrate. LAPs possess complex architectures obtained through unique assemblies of conserved domains associated with lipid, protein and carbohydrate interactions, including the name-defining LCCL domain. Here, we assessed the prevalence of Plasmodium LAP orthologues across eukaryotic life. Our findings show orthologous conservation in all apicomplexans, with lineage-specific repertoires acquired through differential lap gene loss and duplication. Besides Apicomplexa, LAPs are found in their closest relatives: the photosynthetic chromerids, which encode the broadest repertoire including a novel membrane-bound LCCL protein. LAPs are notably absent from other alveolate lineages (dinoflagellates, perkinsids and ciliates), but are encoded by predatory colponemids, a sister group to the alveolates. These results reveal that the LAPs are much older than previously thought and pre-date not only the Apicomplexa but the Alveolata altogether.


Subject(s)
Evolution, Molecular , Phylogeny , Plasmodium , Protozoan Proteins , Protozoan Proteins/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Plasmodium/genetics , Plasmodium/metabolism , Alveolata/genetics , Alveolata/metabolism , Protein Domains , Apicomplexa/genetics , Apicomplexa/metabolism , Lectins/genetics , Lectins/metabolism , Lectins/chemistry
4.
Nat Commun ; 15(1): 4697, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824128

ABSTRACT

Differentiation of male gametocytes into flagellated fertile male gametes relies on the assembly of axoneme, a major component of male development for mosquito transmission of the malaria parasite. RNA-binding protein (RBP)-mediated post-transcriptional regulation of mRNA plays important roles in eukaryotic sexual development, including the development of female Plasmodium. However, the role of RBP in defining the Plasmodium male transcriptome and its function in male gametogenesis remains incompletely understood. Here, we performed genome-wide screening for gender-specific RBPs and identified an undescribed male-specific RBP gene Rbpm1 in the Plasmodium. RBPm1 is localized in the nucleus of male gametocytes. RBPm1-deficient parasites fail to assemble the axoneme for male gametogenesis and thus mosquito transmission. RBPm1 interacts with the spliceosome E complex and regulates the splicing initiation of certain introns in a group of 26 axonemal genes. RBPm1 deficiency results in intron retention and protein loss of these axonemal genes. Intron deletion restores axonemal protein expression and partially rectifies axonemal defects in RBPm1-null gametocytes. Further splicing assays in both reporter and endogenous genes exhibit stringent recognition of the axonemal introns by RBPm1. The splicing activator RBPm1 and its target introns constitute an axonemal intron splicing program in the post-transcriptional regulation essential for Plasmodium male development.


Subject(s)
Axoneme , Introns , Protozoan Proteins , RNA Splicing , RNA-Binding Proteins , Introns/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Animals , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Male , Axoneme/metabolism , Female , Gametogenesis/genetics , Spliceosomes/metabolism , Spliceosomes/genetics , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Malaria/parasitology , Plasmodium/genetics , Plasmodium/metabolism
5.
Int J Mol Sci ; 25(11)2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38892332

ABSTRACT

Post-translational modifications (PTMs) are essential for regulating protein functions, influencing various fundamental processes in eukaryotes. These include, but are not limited to, cell signaling, protein trafficking, the epigenetic control of gene expression, and control of the cell cycle, as well as cell proliferation, differentiation, and interactions between cells. In this review, we discuss protein PTMs that play a key role in the malaria parasite biology and its pathogenesis. Phosphorylation, acetylation, methylation, lipidation and lipoxidation, glycosylation, ubiquitination and sumoylation, nitrosylation and glutathionylation, all of which occur in malarial parasites, are reviewed. We provide information regarding the biological significance of these modifications along all phases of the complex life cycle of Plasmodium spp. Importantly, not only the parasite, but also the host and vector protein PTMs are often crucial for parasite growth and development. In addition to metabolic regulations, protein PTMs can result in epitopes that are able to elicit both innate and adaptive immune responses of the host or vector. We discuss some existing and prospective results from antimalarial drug discovery trials that target various PTM-related processes in the parasite or host.


Subject(s)
Life Cycle Stages , Plasmodium , Protein Processing, Post-Translational , Protozoan Proteins , Humans , Animals , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Plasmodium/metabolism , Plasmodium/genetics , Malaria/parasitology , Malaria/metabolism , Host-Parasite Interactions
6.
Parasit Vectors ; 17(1): 261, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886827

ABSTRACT

BACKGROUND: Malaria transmission in Tanzania is driven by mosquitoes of the Anopheles gambiae complex and Anopheles funestus group. The latter includes An. funestus s.s., an anthropophilic vector, which is now strongly resistant to public health insecticides, and several sibling species, which remain largely understudied despite their potential as secondary vectors. This paper provides the initial results of a cross-country study of the species composition, distribution and malaria transmission potential of members of the Anopheles funestus group in Tanzania. METHODS: Mosquitoes were collected inside homes in 12 regions across Tanzania between 2018 and 2022 using Centres for Disease Control and Prevention (CDC) light traps and Prokopack aspirators. Polymerase chain reaction (PCR) assays targeting the noncoding internal transcribed spacer 2 (ITS2) and 18S ribosomal DNA (18S rDNA) were used to identify sibling species in the An. funestus group and presence of Plasmodium infections, respectively. Where DNA fragments failed to amplify during PCR, we sequenced the ITS2 region to identify any polymorphisms. RESULTS: The following sibling species of the An. funestus group were found across Tanzania: An. funestus s.s. (50.3%), An. parensis (11.4%), An. rivulorum (1.1%), An. leesoni (0.3%). Sequencing of the ITS2 region in the nonamplified samples showed that polymorphisms at the priming sites of standard species-specific primers obstructed PCR amplification, although the ITS2 sequences closely matched those of An. funestus s.s., barring these polymorphisms. Of the 914 samples tested for Plasmodium infections, 11 An. funestus s.s. (1.2%), and 2 An. parensis (0.2%) individuals were confirmed positive for P. falciparum. The highest malaria transmission intensities [entomological inoculation rate (EIR)] contributed by the Funestus group were in the north-western region [108.3 infectious bites/person/year (ib/p/y)] and the south-eastern region (72.2 ib/p/y). CONCLUSIONS: Whereas An. funestus s.s. is the dominant malaria vector in the Funestus group in Tanzania, this survey confirms the occurrence of Plasmodium-infected An. parensis, an observation previously made in at least two other occasions in the country. The findings indicate the need to better understand the ecology and vectorial capacity of this and other secondary malaria vectors in the region to improve malaria control.


Subject(s)
Anopheles , Malaria , Mosquito Vectors , Anopheles/genetics , Anopheles/classification , Anopheles/parasitology , Anopheles/physiology , Animals , Tanzania/epidemiology , Mosquito Vectors/genetics , Mosquito Vectors/parasitology , Mosquito Vectors/classification , Mosquito Vectors/physiology , Malaria/transmission , Malaria/epidemiology , Humans , RNA, Ribosomal, 18S/genetics , Polymerase Chain Reaction , Female , Plasmodium/genetics , Plasmodium/isolation & purification , Plasmodium/classification , DNA, Ribosomal Spacer/genetics
7.
Anal Chem ; 96(19): 7524-7531, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38695755

ABSTRACT

Asymptomatic infections of Plasmodium parasites are major obstacles to malaria control and elimination. A sensitive, specific, and user-friendly method is urgently needed for point-of-care (POC) Plasmodium diagnostics in asymptomatic malaria, especially in resource-limited settings. In this work, we present a POC method (termed Cas13a-SDT) based on the cascade sequence recognition and signal amplification of dual Cas13a trans-cleavage and strand displacement-triggered transcription (SDT). Cas13a-SDT not only achieves exceptional specificity in discriminating the target RNA from nontarget RNAs with any cross-interaction but also meets the sensitivity criterion set by the World Health Organization (WHO) for effective malaria detection. Remarkably, this novel method was successfully applied to screen malaria in asymptomatic infections from clinical samples. The proposed method provides a user-friendly and visually interpretable output mode while maintaining high accuracy and reliability comparable to RT-PCR. These excellent features demonstrate the significant potential of Cas13a-SDT for POC diagnosis of Plasmodium infections, laying a vital foundation for advancing malaria control and elimination efforts.


Subject(s)
CRISPR-Cas Systems , Malaria , Point-of-Care Systems , Malaria/diagnosis , Malaria/parasitology , Humans , CRISPR-Cas Systems/genetics , Plasmodium/genetics , Plasmodium/isolation & purification , Transcription, Genetic
8.
Acta Trop ; 255: 107249, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38740319

ABSTRACT

BACKGROUND: Natural human infections by Plasmodium cynomolgi and P. inui have been reported recently and gain the substantial attention from Southeast Asian countries. Zoonotic transmission of non-human malaria parasites to humans from macaque monkeys occurred through the bites of the infected mosquitoes. The objective of this study is to establish real-time fluorescence loop-mediated isothermal amplification (LAMP) assays for the detection of zoonotic malaria parasites by combining real-time fluorescent technology with the isothermal amplification technique. METHODS: By using 18S rRNA as the target gene, the primers for P. cynomolgi, P. coatneyi and P. inui were newly designed in the present study. Four novel real-time fluorescence LAMP assays were developed for the detection of P. cynomolgi, P. coatneyi, P. inui and P. knowlesi. The entire amplification process was completed in 60 min, with the assays performed at 65 °C. By using SYTO-9 as the nucleic acid intercalating dye, the reaction was monitored via real-time fluorescence signal. RESULTS: There was no observed cross-reactivity among the primers from different species. All 70 field-collected monkey samples were successfully amplified by real-time fluorescence LAMP assays. The detection limit for P. cynomolgi, P. coatneyi and P. knowlesi was 5 × 109 copies/µL. Meanwhile, the detection limit of P. inui was 5 × 1010 copies/µL. CONCLUSION: This is the first report of the detection of four zoonotic malaria parasites by real-time fluorescence LAMP approaches. It is an effective, rapid and simple-to-use technique. This presented platform exhibits considerable potential as an alternative detection for zoonotic malaria parasites.


Subject(s)
Malaria , Molecular Diagnostic Techniques , Nucleic Acid Amplification Techniques , Plasmodium , RNA, Ribosomal, 18S , Sensitivity and Specificity , Zoonoses , Animals , Nucleic Acid Amplification Techniques/methods , Malaria/diagnosis , Malaria/parasitology , Malaria/veterinary , RNA, Ribosomal, 18S/genetics , Molecular Diagnostic Techniques/methods , Plasmodium/genetics , Plasmodium/isolation & purification , Plasmodium/classification , Zoonoses/parasitology , Zoonoses/diagnosis , Humans , DNA Primers/genetics , Fluorescence , Macaca/parasitology , Monkey Diseases/parasitology , Monkey Diseases/diagnosis
9.
Science ; 384(6695): 513-514, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38696584

ABSTRACT

Natural infections are distinct from those of laboratory-or zombie-strains.


Subject(s)
Malaria , Plasmodium , Animals , Humans , Malaria/parasitology , Plasmodium/genetics , Plasmodium/growth & development
10.
PLoS One ; 19(5): e0304179, 2024.
Article in English | MEDLINE | ID: mdl-38820542

ABSTRACT

This study investigates the molecular prevalence and phylogenetic characteristics of two prominent blood-borne pathogens, Toxoplasma gondii (T. gondii) and Plasmodium spp., in common quails (Coturnix coturnix) sampled from both wild (N = 236) and farmed (N = 197) populations across four districts (Layyah, Dera Ghazi Khan, Lahore, and Multan) in Punjab, Pakistan, during the hunting seasons from 2021 to 2023. Additionally, the impact of these pathogens on the complete blood count (CBC) of the hosts is examined. Out of 433 quails tested, 25 (5.8%) exhibited amplification of the internal transcribed spacer (ITS-1) gene for T. gondii, while 15 (3.5%) showed amplification of the Cytochrome b gene for Plasmodium spp. A risk factor analysis indicated that the prevalence of both pathogens was not confined to specific sampling sites or bird sexes (P > 0.05). District-wise analysis highlighted that hens were more susceptible to both T. gondii and Plasmodium spp. infections than cocks. Wild quails exhibited a higher susceptibility to T. gondii compared to farmed birds. Significant CBC variations were recorded in infected birds as compared to uninfected ones. BLAST analysis of generated sequences has confirmed the identity of recovered PCR amplicons as T. gondii and Plasmodium relictum. Phylogenetic analysis revealed that Pakistani isolates clustered with those reported from various countries globally. This study provides the first documentation of T. gondii and Plasmodium sp. infections in Pakistani quails, underscoring the need for detailed investigations across different regions to enhance our understanding of infection rates and the zoonotic potential of these parasites.


Subject(s)
Phylogeny , Plasmodium , Toxoplasma , Toxoplasmosis, Animal , Animals , Pakistan/epidemiology , Toxoplasma/genetics , Plasmodium/genetics , Plasmodium/isolation & purification , Plasmodium/classification , Prevalence , Toxoplasmosis, Animal/epidemiology , Toxoplasmosis, Animal/parasitology , Coturnix/parasitology , Female , Malaria, Avian/epidemiology , Malaria, Avian/parasitology , Male , Poultry Diseases/parasitology , Poultry Diseases/epidemiology
11.
Acta Trop ; 256: 107260, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38782110

ABSTRACT

Avian haemosporidian parasites are spread worldwide and pose a threat to their hosts occasionally. A complete life cycle of these parasites requires two hosts: vertebrate and invertebrate (a blood-sucking insect that acts as a vector). In this study, we tested wild-caught mosquitoes for haemosporidian infections. Mosquitoes were collected (2021-2023) in several localities in Lithuania using a sweeping net and a CDC trap baited with CO2, morphologically identified, and preparations of salivary glands were prepared (from females collected in 2022-2023). 2093 DNA samples from either individual after dissection (1675) or pools (418 pools/1145 individuals) of female mosquito's abdomens were screened using PCR for the detection of haemosporidian parasite DNA. Salivary gland preparations were analyzed microscopically from each PCR-positive mosquito caught in 2022 and 2023. The average prevalence of haemosporidian parasites for all analyzed samples was 2.0 % and varied between 0.6 % (2021) and 3.5 % (2022). DNA of Plasmodium ashfordi (cytochrome b genetic lineage pGRW02), P. circumflexum (pTURDUS1), P. homonucleophilum (pSW2), P. matutinum (pLINN1), P. vaughani (pSYAT05), Haemoproteus brachiatus (hLK03), H. majoris (hWW2), and H. minutus (hTUPHI01) were detected in mosquitoes. Coquilletidia richiardii (3.5 %) and Culex pipiens (2.9 %) were mosquito species with the highest prevalence of haemosporidian parasite DNA detected. Mixed infections were detected in 16 mosquitoes. In one of the samples, sporozoites of P. matutinum (pLINN1) were found in the salivary gland preparation of Culex pipiens, confirming this mosquito species as a competent vector of Plasmodium matutinum and adding it to the list of the natural vectors of this avian parasite.


Subject(s)
Mosquito Vectors , Plasmodium , Salivary Glands , Animals , Female , Mosquito Vectors/parasitology , Plasmodium/isolation & purification , Plasmodium/genetics , Plasmodium/classification , Salivary Glands/parasitology , Lithuania , Haemosporida/genetics , Haemosporida/isolation & purification , Haemosporida/classification , Culicidae/parasitology , Birds/parasitology , Polymerase Chain Reaction , Culex/parasitology , DNA, Protozoan/genetics
12.
Sci Rep ; 14(1): 9871, 2024 04 30.
Article in English | MEDLINE | ID: mdl-38684775

ABSTRACT

The Plasmodium is responsible for malaria which poses a major health threat, globally. This study is based on the estimation of the relative abundance of mosquitoes, and finding out the correlations of meteorological parameters (temperature, humidity and rainfall) with the abundance of mosquitoes. In addition, this study also focused on the use of nested PCR (species-specific nucleotide sequences of 18S rRNA genes) to explore the Plasmodium spp. in female Anopheles. In the current study, the percentage relative abundance of Culex mosquitoes was 57.65% and Anopheles 42.34% among the study areas. In addition, the highest number of mosquitoes was found in March in district Mandi Bahauddin at 21 °C (Tmax = 27, Tmin = 15) average temperature, 69% average relative humidity and 131 mm rainfall, and these climatic factors were found to affect the abundance of the mosquitoes, directly or indirectly. Molecular analysis showed that overall, 41.3% of the female Anopheles pools were positive for genus Plasmodium. Among species, the prevalence of Plasmodium (P.) vivax (78.1%) was significantly higher than P. falciparum (21.9%). This study will be helpful in the estimation of future risk of mosquito-borne diseases along with population dynamic of mosquitoes to enhance the effectiveness of vector surveillance and control programs.


Subject(s)
Anopheles , Malaria , Mosquito Vectors , Plasmodium , Polymerase Chain Reaction , Animals , Anopheles/parasitology , Anopheles/genetics , Mosquito Vectors/parasitology , Mosquito Vectors/genetics , Polymerase Chain Reaction/methods , Female , Plasmodium/genetics , Plasmodium/isolation & purification , Malaria/epidemiology , Malaria/parasitology , Malaria/transmission , RNA, Ribosomal, 18S/genetics , Culex/parasitology , Culex/genetics , Humans , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Plasmodium vivax/genetics
13.
Acta Trop ; 254: 107187, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38518834

ABSTRACT

Over the past year, P. falciparum infections have declined in Thailand, yet nonhuman primate malaria infections have correspondingly increased, including Plasmodium knowlesi and P. cynomolgi. Nevertheless, little is known about simian malaria in its natural macaque hosts, Macaca mulatta and Macaca fascicularis. This study aims to address several research questions, including the prevalence and distribution of simian malaria in these two Thai wild macaque species, variations in infection between different macaque species and between M. fascicularis subspecies, and the genetic composition of these pathogens. Blood samples were collected from 82 M. mulatta and 690 M. fascicularis across 15 locations in Thailand, as well as two locations in Vietnam and Myanmar. We employed quantitative real-time PCR targeting the Plasmodium genus-specific 18S ribosomal RNA (rRNA) gene to detect malaria infection, with a limit of detection set at 1,215.98 parasites per mL. We genotyped eight microsatellite markers, and the P. cynomolgi dihydrofolate reductase gene (DHFR) was sequenced (N = 29). In total, 100 of 772 samples (13 %) tested positive for malaria, including 45 (13 %) for P. cynomolgi, 37 (13 %) for P. inui, 16 (5 %) for P. coatneyi, and 2 (0.25 %) for Hepatocystis sp. in Saraburi, central and Ranong, southern Thailand. Notably, simian malaria infection was observed exclusively in M. fascicularis and not in M. mulatta (P = 0.0002). Particularly, P. cynomolgi was detected in 21.7 % (45/207) of M. f. fascicularis living in Wat Tham Phrapothisat, Saraburi Province. The infection with simian malaria was statistically different between M. fascicularis and M. mulatta (P = 0.0002) but not within M. fascicularis subspecies (P = 0.78). A haplotype network analysis revealed that P. cynomolgi shares a lineage with reference strains obtained from macaques. No mutation in the predicted binding pocket of PcyDHFR to pyrimethamine was observed. This study reveals a significant prevalence of simian malaria infection in M. fascicularis. The clonal genotypes of P. cynomolgi suggest in-reservoir breeding. These findings raise concerns about the potential spread of nonhuman primate malaria to humans and underscore the need for preventive measures.


Subject(s)
Genetic Variation , Macaca fascicularis , Malaria , RNA, Ribosomal, 18S , Animals , Thailand/epidemiology , Malaria/epidemiology , Malaria/parasitology , Malaria/veterinary , Macaca fascicularis/parasitology , Prevalence , RNA, Ribosomal, 18S/genetics , Macaca mulatta/parasitology , Genotype , Microsatellite Repeats/genetics , Monkey Diseases/parasitology , Monkey Diseases/epidemiology , Humans , Myanmar/epidemiology , Tetrahydrofolate Dehydrogenase/genetics , Plasmodium knowlesi/genetics , Plasmodium knowlesi/isolation & purification , Plasmodium/genetics , Plasmodium/classification , Plasmodium/isolation & purification , Vietnam/epidemiology , DNA, Protozoan/genetics , Plasmodium cynomolgi/genetics , Plasmodium cynomolgi/classification , Real-Time Polymerase Chain Reaction
14.
Acta Trop ; 254: 107191, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38554994

ABSTRACT

Malaria remains one of the most perilous vector-borne infectious diseases for humans globally. Sexual gametocyte represents the exclusive stage at which malaria parasites are transmitted from the vertebrate to the Anopheles host. The feasible and effective approach to prevent malaria transmission is by addressing the sexual developmental processes, that is, gametocytogenesis and gametogenesis. Thus, this review will comprehensively cover advances in the regulation of gene expression surrounding the transmissible stages, including epigenetic, transcriptional, and post-transcriptional control.


Subject(s)
Anopheles , Plasmodium , Animals , Anopheles/parasitology , Anopheles/genetics , Plasmodium/genetics , Plasmodium/growth & development , Mosquito Vectors/genetics , Mosquito Vectors/parasitology , Mosquito Vectors/growth & development , Gametogenesis/genetics , Humans , Malaria/transmission , Malaria/parasitology , Gene Expression Regulation , Gene Expression Regulation, Developmental , Epigenesis, Genetic , Sexual Development/genetics
15.
Parasit Vectors ; 17(1): 150, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38519966

ABSTRACT

BACKGROUND: Mosquitoes (Culicidae) are vectors for most malaria parasites of the Plasmodium species and are required for Plasmodium spp. to complete their life cycle. Despite having 16 species of mosquitoes and the detection of many Plasmodium species in birds, little is known about the role of different mosquito species in the avian malaria life cycle in New Zealand. METHODS: In this study, we used nested polymerase chain reaction (PCR) and real-time PCR to determine Plasmodium spp. prevalence and diversity of mitochondrial cytochrome b gene sequences in wild-caught mosquitoes sampled across ten sites on the North Island of New Zealand during 2012-2014. The mosquitoes were pooled by species and location collected, and the thorax and abdomens were examined separately for Plasmodium spp. DNA. Akaike information criterion (AIC) modeling was used to test whether location, year of sampling, and mosquito species were significant predictors of minimum infection rates (MIR). RESULTS: We collected 788 unengorged mosquitoes of six species, both native and introduced. The most frequently caught mosquito species were the introduced Aedes notoscriptus and the native Culex pervigilans. Plasmodium sp DNA was detected in 37% of matched thorax and abdomen pools. When considered separately, 33% of abdomen and 23% of thorax pools tested positive by nested PCR. The MIR of the positive thorax pools from introduced mosquito species was 1.79% for Ae. notoscriptus and 0% for Cx. quinquefasciatus, while the MIR for the positive thorax pools of native mosquito species was 4.9% for Cx. pervigilans and 0% for Opifex fuscus. For the overall MIR, site and mosquito species were significant predictors of Plasmodium overall MIR. Aedes notoscriptus and Cx. pervigilans were positive for malaria DNA in the thorax samples, indicating that they may play a role as avian malaria vectors. Four different Plasmodium lineages (SYAT05, LINN1, GRW6, and a new lineage of P (Haemamoeba) sp. AENOT11) were identified in the pooled samples. CONCLUSIONS: This is the first detection of avian Plasmodium DNA extracted from thoraxes of native Culex and introduced Aedes mosquito species in New Zealand and therefore the first study providing an indication of potential vectors in this country.


Subject(s)
Aedes , Anopheles , Culex , Malaria, Avian , Malaria , Plasmodium , Animals , Malaria, Avian/parasitology , Anopheles/genetics , New Zealand/epidemiology , Mosquito Vectors/parasitology , Culex/genetics , Plasmodium/genetics , Aedes/genetics , Birds/parasitology , DNA, Protozoan/genetics , DNA, Protozoan/analysis
16.
Acta Parasitol ; 69(1): 1000-1004, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38498250

ABSTRACT

BACKGROUND: Prompt malarial treatment and surveillance is crucial for accurate diagnosis of Plasmodium Sp. Gold standard microscopic examination has been widely applied for diagnosis of malaria in most part of the endemic areas. But in case of submicroscopic and asymptomatic microscopic diagnosis is questioned. The study aims to develop a simple, cost effective & robust nucleic acid amplification technique for the detection of malaria parasite. METHODS: Study population included 50 clinically diagnosed positive malaria patient samples from various pathological laboratories. Microscopy by preparing thick film was carried out of every sample for primary screening in the available facility of Surat Raktadan Kendra & Research Centre- Blood Bank. The conventional PCR (Polymerase Chain Reaction) was applied for genus-specific amplification targeting the 18 S rRNA gene of Plasmodium. Agarose gel electrophoresis was used to separate and analyze the amplified PCR product using 2% Agarose gel. RESULTS AND CONCLUSION: The study shows that nested PCR not only detected all microscopic positive samples, but also detected submicroscopic infections that were missed or misread by microscopy. Hence, the sensitivity of molecular based detection technique is proved to be more compared to microscopic examination.


Subject(s)
Malaria , Polymerase Chain Reaction , RNA, Ribosomal, 18S , Sensitivity and Specificity , Humans , Malaria/diagnosis , Malaria/parasitology , Polymerase Chain Reaction/methods , RNA, Ribosomal, 18S/genetics , Plasmodium/genetics , Plasmodium/isolation & purification , Plasmodium/classification , Molecular Diagnostic Techniques/methods , Nucleic Acid Amplification Techniques/methods , Microscopy/methods , DNA, Protozoan/genetics
17.
Parasitol Int ; 101: 102891, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38537686

ABSTRACT

Malaria remains a significant global public health concern, with a recent increase in the number of zoonotic malaria cases in Southeast Asian countries. However, limited reports on the vector for zoonotic malaria exist owing to difficulties in detecting parasite DNA in Anopheles mosquito vectors. Herein, we demonstrate for the first time that several Anopheles mosquitoes contain simian malaria parasite DNA using droplet digital PCR (ddPCR), a highly sensitive PCR method. An entomological survey was conducted to identify simian malaria vector species at Phra Phothisat Temple (PPT), central Thailand, recognized for a high prevalence of simian malaria in wild cynomolgus macaques. A total of 152 mosquitoes from six anopheline species were collected and first analyzed by a standard 18S rRNA nested-PCR analysis for malaria parasite which yielded negative results in all collected mosquitoes. Later, ddPCR was used and could detect simian malaria parasite DNA, i.e. Plasmodium cynomolgi, in 25 collected mosquitoes. And this is the first report of simian malaria parasite DNA detection in Anopheles sawadwongporni. This finding proves that ddPCR is a powerful tool for detecting simian malarial parasite DNA in Anopheles mosquitoes and can expand our understanding of the zoonotic potential of malaria transmission between monkeys and humans.


Subject(s)
Anopheles , Malaria , Mosquito Vectors , Polymerase Chain Reaction , Anopheles/parasitology , Animals , Polymerase Chain Reaction/methods , Malaria/transmission , Malaria/epidemiology , Malaria/parasitology , Malaria/diagnosis , Mosquito Vectors/parasitology , Thailand/epidemiology , RNA, Ribosomal, 18S/analysis , RNA, Ribosomal, 18S/genetics , Plasmodium/isolation & purification , Plasmodium/genetics , Macaca fascicularis/parasitology , DNA, Protozoan/analysis , Humans , Sensitivity and Specificity
18.
Mol Ecol ; 33(8): e17329, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38533805

ABSTRACT

Patterns of pathogen prevalence are, at least partially, the result of coevolutionary host-pathogen interactions. Thus, exploring the distribution of host genetic variation in relation to infection by a pathogen within and across populations can provide important insights into mechanisms of host defence and adaptation. Here, we use a landscape genomics approach (Bayenv) in conjunction with genome-wide data (ddRADseq) to test for associations between avian malaria (Plasmodium) prevalence and host genetic variation across 13 populations of the island endemic Berthelot's pipit (Anthus berthelotii). Considerable and consistent spatial heterogeneity in malaria prevalence was observed among populations over a period of 15 years. The prevalence of malaria infection was also strongly positively correlated with pox (Avipoxvirus) prevalence. Multiple host loci showed significant associations with malaria prevalence after controlling for genome-wide neutral genetic structure. These sites were located near to or within genes linked to metabolism, stress response, transcriptional regulation, complement activity and the inflammatory response, many previously implicated in vertebrate responses to malarial infection. Our findings identify diverse genes - not just limited to the immune system - that may be involved in host protection against malaria and suggest that spatially variable pathogen pressure may be an important evolutionary driver of genetic divergence among wild animal populations, such as Berthelot's pipit. Furthermore, our data indicate that spatio-temporal variation in multiple different pathogens (e.g. malaria and pox in this case) may have to be studied together to develop a more holistic understanding of host pathogen-mediated evolution.


Subject(s)
Malaria, Avian , Passeriformes , Plasmodium , Animals , Malaria, Avian/epidemiology , Malaria, Avian/genetics , Plasmodium/genetics , Genetic Drift , Passeriformes/genetics , Genotype
19.
Malar J ; 23(1): 70, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38459560

ABSTRACT

BACKGROUND: Birds chronically infected with avian malaria parasites often show relapses of parasitaemia after latent stages marked by absence of parasites in the peripheral circulation. These relapses are assumed to result from the activation of dormant exo-erythrocytic stages produced during secondary (post-erythrocytic) merogony of avian Plasmodium spp. Yet, there is no morphological proof of persistent or dormant tissue stages in the avian host during latent infections. This study investigated persistence of Plasmodium relictum pSGS1 in birds with latent infections during winter, with the goal to detect presumed persisting tissue stages using a highly sensitive RNAscope® in situ hybridization technology. METHODS: Fourteen domestic canaries were infected with P. relictum pSGS1 by blood-inoculation in spring, and blood films examined during the first 4 months post infection, and during winter and spring of the following year. After parasitaemia was no longer detectable, half of the birds were dissected, and tissue samples investigated for persisting tissue stages using RNAscope ISH and histology. The remaining birds were blood-checked and dissected after re-appearance of parasitaemia, and their tissues equally examined. RESULTS: Systematic examination of tissues showed no exo-erythrocytic stages in birds exhibiting latent infections by blood-film microscopy, indicating absence of dormant tissue stages in P. relictum pSGS1-infected canaries. Instead, RNAscope ISH revealed rare P. relictum blood stages in capillaries of various tissues and organs, demonstrating persistence of the parasites in the microvasculature. Birds examined after re-appearance of parasitemia showed higher numbers of P. relictum blood stages in both capillaries and larger blood vessels, indicating replication during early spring and re-appearance in the peripheral circulation. CONCLUSIONS: The findings suggest that persistence of P. relictum pSGS1 during latent infection is mediated by continuous low-level erythrocytic merogony and possibly tissue sequestration of infected blood cells. Re-appearance of parasitaemia in spring seems to result from increased erythrocytic merogony, therefore representing recrudescence and not relapse in blood-inoculated canaries. Further, the study highlights strengths and limitations of the RNAscope ISH technology for the detection of rare parasite stages in tissues, providing directions for future research on persistence and tissue sequestration of avian malaria and related haemosporidian parasites.


Subject(s)
Latent Infection , Malaria, Avian , Plasmodium , Animals , Canaries/parasitology , Malaria, Avian/parasitology , Plasmodium/genetics , Birds , In Situ Hybridization , Parasitemia/parasitology , Recurrence
20.
Parasit Vectors ; 17(1): 104, 2024 Mar 02.
Article in English | MEDLINE | ID: mdl-38431686

ABSTRACT

BACKGROUND: Mosquitoes belonging to the Anopheles gambiae sensu lato complex play a major role in malaria transmission across Africa. This study assessed the relative importance of members of An. gambiae s.l. in malaria transmission in two rural villages in the Republic of the Congo. METHODS: Adult mosquitoes were collected using electric aspirators from June to September 2022 in Djoumouna and Ntoula villages and were sorted by taxa based on their morphological features. Anopheles gambiae s.l. females were also molecularly identified. A TaqMan-based assay and a nested polymerase chain reaction (PCR) were performed to determine Plasmodium spp. in the mosquitoes. Entomological indexes were estimated, including man-biting rate, entomological inoculation rate (EIR), and diversity index. RESULTS: Among 176 mosquitoes collected, An. gambiae s.l. was predominant (85.8%), followed by Culex spp. (13.6%) and Aedes spp. (0.6%). Three members of the An. gambiae s.l. complex were collected in both villages, namely An. gambiae sensu stricto (74.3%), Anopheles coluzzii (22.9%) and Anopheles arabiensis (2.8%). Three Plasmodium species were detected in An. gambiae s.s. and An. coluzzii (Plasmodium falciparum, P. malariae and P. ovale), while only P. falciparum and P. malariae were found in An. arabiensis. In general, the Plasmodium infection rate was 35.1% (53/151) using the TaqMan-based assay, and nested PCR confirmed 77.4% (41/53) of those infections. The nightly EIR of An. gambiae s.l. was 0.125 infectious bites per person per night (ib/p/n) in Djoumouna and 0.08 ib/p/n in Ntoula. The EIR of An. gambiae s.s. in Djoumouna (0.11 ib/p/n) and Ntoula (0.04 ib/p/n) was higher than that of An. coluzzii (0.01 and 0.03 ib/p/n) and An. arabiensis (0.005 and 0.0 ib/p/n). CONCLUSIONS: This study provides baseline information on the dominant vectors and dynamics of malaria transmission in the rural areas of the Republic of the Congo during the dry season. In the two sampled villages, An. gambiae s.s. appears to play a predominant role in Plasmodium spp.


Subject(s)
Anopheles , Malaria, Falciparum , Malaria , Plasmodium , Humans , Male , Animals , Female , Seasons , Congo/epidemiology , Mosquito Vectors , Malaria/epidemiology , Plasmodium/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...