Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.352
Filter
1.
PLoS One ; 19(7): e0306289, 2024.
Article in English | MEDLINE | ID: mdl-38950022

ABSTRACT

Although the overall burden of malaria is decreasing in Ethiopia, a recent report of an unpredictable increased incidence may be related to the presence of community-wide gametocyte-carrier individuals and a high proportion of infected vectors. This study aimed to reveal the current prevalence of gametocyte-carriage and the sporozoite infectivity rate of Anopheles vectors for Plasmodium parasites. A community-based cross-sectional study was conducted from May 01 to June 30/2019. A total of 53 households were selected using systematic random sampling and a 242 study participants were recruited. Additionally,515 adult female Anopheles mosquitoes were collected using Center for Diseases Control and Prevention (CDC) light traps and mouth aspirators. Parasite gametocytemia was determined using giemsa stain microscopy, while sporozoite infection was determined by giemsa staining microscopy and enzyme linked immunosorbent assay (ELISA). Among the total 242 study participants, 5.4% (95%, CI = 2.9-8.3) of them were positive for any of the Plasmodium species gametocyte. Furthermore, being female [AOR = 15.5(95%, CI = 1.71-140.39)], age group between 15-29 years old [AOR = 16.914 (95%, CI = 1.781-160.63)], no ITNs utilization [AOR = 16.7(95%, CI = 1.902 -146.727)], and high asexual parasite density [(95%, CI = 0.057-0.176, P = 0.001, F = 18.402)] were identified as statistically significant factors for gametocyte carriage. Whereas sporozoite infection rate was 11.6% (95%, CI = 8.2-15.5) and 12.7% (95%, CI = 9.6-16.3) by microscopy and ELISA, respectively. Overall, this study indicated that malaria remains to be an important public health problem in Gondar Zuria district where high gametocyte carriage rate and sporozoite infection rate could sustain its transmission and burden. Therefore, in Ethiopia, where malaria elimination program is underway, frequent, and active community-based surveillance of gametocytemia and sporozoite infection rate is important.


Subject(s)
Anopheles , Mosquito Vectors , Sporozoites , Animals , Ethiopia/epidemiology , Humans , Anopheles/parasitology , Female , Adult , Sporozoites/physiology , Adolescent , Young Adult , Male , Cross-Sectional Studies , Mosquito Vectors/parasitology , Child , Child, Preschool , Malaria/epidemiology , Malaria/parasitology , Malaria/transmission , Middle Aged , Plasmodium/isolation & purification , Infant , Plasmodium falciparum/isolation & purification , Plasmodium falciparum/physiology , Prevalence
2.
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
3.
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
4.
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
5.
Mol Biochem Parasitol ; 259: 111635, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38857772

ABSTRACT

Malaria, a parasitic infection caused by the genus Plasmodium, results to over 20 million reported cases annually worldwide. Most individuals exhibit various symptoms, and blood analysis plays a crucial role in determining the appropriate treatment approach. This study discusses various hematologic complications associated with different Plasmodium species. A review of scientific databases including PubMed, Science Direct, Web of Science, Scopus, EMBASE, Magiran, SID, IranMedex was conducted using standard keywords such as Plasmodium, malaria, anemia and blood disorders (hematologic disorder) between 2000 and 2024. The review focused on articles pertaining to clinical trials, prospective cohort, retrospective, cross-sectional and case-control studies. Articles evaluating the effects of malaria on blood cells and indices, with target groups including human and animals, were included. Articles not written in English or Farsi were excluded. Our review revealed that, apart from iron deficiency anemia and vascular dysfunction contributed in part by adhesion of infected RBC to endothelium, decreases in hematocrit and hemoglobin levels, as part of pancytopenia and thrombocytopenia, are characteristic of Plasmodium infection. Additionally, the occurrence of inflammation due to the release of inflammatory cytokines and complement activation can complicate the clinical features of malaria in individuals with hematologic conditions.


Subject(s)
Malaria , Humans , Malaria/parasitology , Animals , Plasmodium , Hematologic Diseases , Anemia/etiology
6.
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
7.
Expert Rev Vaccines ; 23(1): 645-654, 2024.
Article in English | MEDLINE | ID: mdl-38888098

ABSTRACT

INTRODUCTION: Malaria continues to remain a major global health problem with nearly a quarter of a billion clinical cases and more than 600,000 deaths in 2022. There has been significant progress toward vaccine development, however, poor efficacy of approved vaccines requiring multiple immunizing doses emphasizes the need for continued efforts toward improved vaccines. Progress to date, nonetheless, has provided impetus for malaria elimination. AREAS COVERED: In this review we will focus on diverse immune mechanisms targeting gametocytes in the human host and gametocyte-mediated malaria transmission via the mosquito vector. EXPERT OPINION: To march toward the goal of malaria elimination it will be critical to target the process of malaria transmission by mosquitoes, mediated exclusively by the sexual stages, i.e. male, and female gametocytes, ingested from infected vertebrate host. Studies over several decades have established antigens in the parasite sexual stages developing in the mosquito midgut as attractive targets for the development of transmission blocking vaccines (TBVs). Immune clearance of gametocytes in the vertebrate host can synergize with TBVs and directly aid in maintaining effective transmission reducing immune potential.


Subject(s)
Malaria Vaccines , Malaria , Mosquito Vectors , Vaccine Development , Humans , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Animals , Malaria/prevention & control , Malaria/transmission , Malaria/immunology , Malaria/parasitology , Mosquito Vectors/parasitology , Mosquito Vectors/immunology , Plasmodium/immunology
8.
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
9.
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
10.
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
11.
J Wildl Dis ; 60(3): 634-646, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38741368

ABSTRACT

Pathogens have traditionally been studied in isolation within host systems; yet in natural settings they frequently coexist. This raises questions about the dynamics of co-infections and how host life-history traits might predict co-infection versus single infection. To address these questions, we investigated the presence of two parasites, a gut parasite (Isospora coccidians) and a blood parasite (Plasmodium spp.), in House Finches (Haemorhous mexicanus), a common passerine bird in North America. We then correlated these parasitic infections with various health and condition metrics, including hematological parameters, plasma carotenoids, lipid-soluble vitamins, blood glucose concentration, body condition, and prior disease history. Our study, based on 48 birds captured in Tempe, Arizona, US, in October 2021, revealed that co-infected birds exhibited elevated circulating lutein levels and a higher heterophil:lymphocyte ratio (H/L ratio) compared to those solely infected with coccidia Isospora spp. This suggests that co-infected birds experience heightened stress and may use lutein to bolster immunity against both pathogens, and that there are potentially toxic effects of lutein in co-infected birds compared to those infected solely with coccidia Isospora sp. Our findings underscore the synergistic impact of coparasitism, emphasizing the need for more co-infection studies to enhance our understanding of disease dynamics in nature, as well as its implications for wildlife health and conservation efforts.


Subject(s)
Bird Diseases , Coccidiosis , Coinfection , Finches , Isospora , Malaria, Avian , Plasmodium , Animals , Finches/parasitology , Coinfection/veterinary , Coinfection/parasitology , Coinfection/epidemiology , Malaria, Avian/epidemiology , Malaria, Avian/parasitology , Malaria, Avian/blood , Bird Diseases/parasitology , Bird Diseases/epidemiology , Bird Diseases/blood , Isospora/isolation & purification , Coccidiosis/veterinary , Coccidiosis/epidemiology , Coccidiosis/parasitology , Plasmodium/isolation & purification , Isosporiasis/veterinary , Isosporiasis/epidemiology , Isosporiasis/parasitology , Arizona/epidemiology , Male , Female
12.
Biochem Pharmacol ; 225: 116260, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38705539

ABSTRACT

Malaria is caused by an apicomplexan protozoan parasite, Plasmodium, and is transmitted through vectors. It remains a substantial health burden, especially in developing countries, leading to significant socioeconomic losses. Although the World Health Organization (WHO) has approved various antimalarial medications in the past two decades, the increasing resistance to these medications has worsened the situation. The development of drug resistance stems from genetic diversity among Plasmodium strains, impeding eradication efforts. Consequently, exploring innovative technologies and strategies for developing effective medications based on the host is crucial. Artemisinin and its derivatives (artemisinins) have been recommended by the WHO for treating malaria owing to their known effectiveness in killing the parasite. However, their potential to target the host for malaria treatment has not been investigated. This article concisely reviews the application of host-directed therapeutics, potential drug candidates targeting the host for treating malaria, and usage of artemisinins in numerous diseases. It underscores the importance of host-directed interventions for individuals susceptible to malaria, suggests the potential utility of artemisinins in host-directed malaria treatments, and posits that the modulation of host proteins with artemisinins may offer a means of intervening in host-parasite interactions. Further studies focusing on the host-targeting perspective of artemisinins can provide new insights into the mechanisms of artemisinin resistance and offer a unique opportunity for new antimalarial drug discovery.


Subject(s)
Antimalarials , Artemisinins , Malaria , Artemisinins/pharmacology , Artemisinins/therapeutic use , Humans , Antimalarials/pharmacology , Antimalarials/therapeutic use , Malaria/drug therapy , Animals , Drug Resistance/drug effects , Plasmodium/drug effects , Host-Parasite Interactions/drug effects
13.
Trends Parasitol ; 40(6): 466-476, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38714463

ABSTRACT

The Plasmodium parasites that cause malaria undergo asymptomatic development in the parenchymal cells of the liver, the hepatocytes, prior to infecting erythrocytes and causing clinical disease. Traditionally, hepatocytes have been perceived as passive bystanders that allow hepatotropic pathogens such as Plasmodium to develop relatively unchallenged. However, now there is emerging evidence suggesting that hepatocytes can mount robust cell-autonomous immune responses that target Plasmodium, limiting its progression to the blood and reducing the incidence and severity of clinical malaria. Here we discuss our current understanding of hepatocyte cell-intrinsic immune responses that target Plasmodium and how these pathways impact malaria.


Subject(s)
Hepatocytes , Malaria , Plasmodium , Plasmodium/immunology , Plasmodium/physiology , Humans , Malaria/immunology , Malaria/parasitology , Hepatocytes/parasitology , Hepatocytes/immunology , Animals
14.
Science ; 384(6696): 697-703, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38723080

ABSTRACT

Changes in climate shift the geographic locations that are suitable for malaria transmission because of the thermal constraints on vector Anopheles mosquitos and Plasmodium spp. malaria parasites and the lack of availability of surface water for vector breeding. Previous Africa-wide assessments have tended to solely represent surface water using precipitation, ignoring many important hydrological processes. Here, we applied a validated and weighted ensemble of global hydrological and climate models to estimate present and future areas of hydroclimatic suitability for malaria transmission. With explicit surface water representation, we predict a net decrease in areas suitable for malaria transmission from 2025 onward, greater sensitivity to future greenhouse gas emissions, and different, more complex, malaria transmission patterns. Areas of malaria transmission that are projected to change are smaller than those estimated by precipitation-based estimates but are associated with greater changes in transmission season lengths.


Subject(s)
Anopheles , Climate Change , Hydrology , Malaria , Mosquito Vectors , Water , Animals , Humans , Africa/epidemiology , Anopheles/parasitology , Greenhouse Gases/analysis , Malaria/transmission , Mosquito Vectors/parasitology , Rain , Seasons , Water/parasitology , Plasmodium , Epidemiological Models
15.
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
16.
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
17.
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
18.
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
19.
Parasit Vectors ; 17(1): 184, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38600596

ABSTRACT

Human malaria, an ancient tropical disease, is caused by infection with protozoan parasites belonging to the genus Plasmodium and is transmitted by female mosquitoes of the genus Anopheles. Our understanding of human malaria parasites began officially in 1880 with their discovery in the blood of malaria patients by Charles Louis Alphonse Lavéran (1845-1922), a French army officer working in Algeria. A claim for priority was made by Philipp Friedrich Hermann Klencke (1813-1881) in 1843, who wrote a chapter entitled: "Marvellous parallelism between the manifestations of vertigo and the presence of animalcule vacuoles in living blood." We should not lose sight of this old controversy, which is rarely mentioned in historical reviews on malaria.


Subject(s)
Anopheles , Malaria , Parasites , Plasmodium , Animals , Humans , Female , Malaria/parasitology , Algeria/epidemiology
20.
Malar J ; 23(1): 112, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38641572

ABSTRACT

BACKGROUND: In malaria endemic regions of the Peruvian Amazon, rainfall together with river level and breeding site availability drive fluctuating vector mosquito abundance and human malaria cases, leading to temporal heterogeneity. The main variables influencing spatial transmission include location of communities, mosquito behaviour, land use/land cover, and human ecology/behaviour. The main objective was to evaluate seasonal and microgeographic biting behaviour of the malaria vector Nyssorhynchus (or Anopheles) darlingi in Amazonian Peru and to investigate effects of seasonality on malaria transmission. METHODS: We captured mosquitoes from 18:00 to 06:00 h using Human Landing Catch in two riverine (Lupuna, Santa Emilia) and two highway (El Triunfo, Nuevo Horizonte) communities indoors and outdoors from 8 houses per community, during the dry and rainy seasons from February 2016 to January 2017. We then estimated parity rate, daily survival and age of a portion of each collection of Ny. darlingi. All collected specimens of Ny. darlingi were tested for the presence of Plasmodium vivax or Plasmodium falciparum sporozoites using real-time PCR targeting the small subunit of the 18S rRNA. RESULTS: Abundance of Ny. darlingi varied across village, season, and biting behaviour (indoor vs outdoor), and was highly significant between rainy and dry seasons (p < 0.0001). Biting patterns differed, although not significantly, and persisted regardless of season, with peaks in highway communities at ~ 20:00 h in contrast to biting throughout the night (i.e., 18:00-06:00) in riverine communities. Of 3721 Ny. darlingi tested for Plasmodium, 23 (0.62%) were infected. We detected Plasmodium-infected Ny. darlingi in both community types and most (20/23) were captured outdoors during the rainy season; 17/23 before midnight. Seventeen Ny. darlingi were infected with P. vivax, and 6 with P. falciparum. No infected Ny. darlingi were captured during the dry season. Significantly higher rates of parity were detected in Ny. darlingi during the rainy season (average 64.69%) versus the dry season (average 36.91%) and by community, Lupuna, a riverine village, had the highest proportion of parous to nulliparous females during the rainy season. CONCLUSIONS: These data add a seasonal dimension to malaria transmission in peri-Iquitos, providing more evidence that, at least locally, the greatest risk of malaria transmission is outdoors during the rainy season mainly before midnight, irrespective of whether the community was located adjacent to the highway or along the river.


Subject(s)
Anopheles , Bites and Stings , Malaria, Falciparum , Malaria, Vivax , Malaria , Plasmodium , Animals , Female , Humans , Anopheles/genetics , Malaria/epidemiology , Peru/epidemiology , Mosquito Vectors , Malaria, Vivax/epidemiology , Seasons
SELECTION OF CITATIONS
SEARCH DETAIL
...