1
|
Neog S, Vinjamuri SR, Vijayan K, Kumar S, Trivedi V. NDV targets the invasion pathway in malaria parasite through cell surface sialic acid interaction. FASEB J 2024; 38:e23856. [PMID: 39092913 DOI: 10.1096/fj.202400004rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/01/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024]
Abstract
Merozoites utilize sialic acids on the red blood cell (RBC) cell surface to rapidly adhere to and invade the RBCs. Newcastle disease virus (NDV) displays a strong affinity toward membrane-bound sialic acids. Incubation of NDV with the malaria parasites dose-dependently reduces its cellular viability. The antiplasmodial activity of NDV is specific, as incubation with Japanese encephalitis virus, duck enteritis virus, infectious bronchitis virus, and influenza virus did not affect the parasite propagation. Interestingly, NDV is reducing more than 80% invasion when RBCs are pretreated with the virus. Removal of the RBC surface proteins or the NDV coat proteins results in disruption of the virus binding to RBC. It suggests the involvement of specific protein: ligand interaction in virus binding. We established that the virus engages with the parasitized RBCs (PRBCs) through its hemagglutinin neuraminidase (HN) protein by recognizing sialic acid-containing glycoproteins on the cell surface. Blocking of the HN protein with free sialic acid or anti-HN antibodies abolished the virus binding as well as its ability to reduce parasite growth. Interestingly, the purified HN from the virus alone could inhibit the parasite's growth in a dose-dependent manner. NDV binds strongly to knobless murine parasite strain Plasmodium yoelii and restricted the parasite growth in mice. Furthermore, the virus was found to preferentially target the PRBCs compared to normal erythrocytes. Immunolocalization studies reveal that NDV is localized on the plasma membrane as well as weakly inside the PRBC. NDV causes neither any infection nor aggregation of the human RBCs. Our findings suggest that NDV is a potential candidate for developing targeted drug delivery platforms for the Plasmodium-infected RBCs.
Collapse
Affiliation(s)
- Siddharth Neog
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India
| | - Sandeep Reddy Vinjamuri
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Kamalakannan Vijayan
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Sachin Kumar
- Viral Immunology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India
| | - Vishal Trivedi
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, India
| |
Collapse
|
2
|
Muhamad P, Phompradit P, Chaijaroenkul W, Na-Bangchang K. Distribution patterns of molecular markers of antimalarial drug resistance in Plasmodium falciparum isolates on the Thai-Myanmar border during the periods of 1993-1998 and 2002-2008. BMC Genomics 2024; 25:269. [PMID: 38468205 DOI: 10.1186/s12864-023-09814-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 11/17/2023] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Polymorphisms of Plasmodium falciparum chloroquine resistance transporter (pfcrt), Plasmodium falciparum multi-drug resistance 1 (pfmdr1) and Plasmodium falciparum kelch 13-propeller (pfk13) genes are accepted as valid molecular markers of quinoline antimalarials and artemisinins. This study investigated the distribution patterns of these genes in P. falciparum isolates from the areas along the Thai-Myanmar border during the two different periods of antimalarial usage in Thailand. RESULTS Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) were used to detect pfcrt mutations at codons 76, 220, 271, 326, 356, and 371 as well as pfmdr1 mutation at codon 86. The prevalence of pfcrt mutations was markedly high (96.4-99.7%) in samples collected during both periods. The proportions of mutant genotypes (number of mutant/total isolate) at codons 76, 220, 271, 326, 356 and 371 in the isolates collected during 1993-1998 (period 1) compared with 2002-2008 (period 2) were 97.9% (137/140) vs. 97.1% (401/413), 97.9% (140/143) vs. 98.8% (171/173), 97.2% (139/143) vs. 97.1% (333/343), 98.6% (140/142) vs. 99.7% (385/386), 96.4% (134/139) vs. 98.2% (378/385) and 97.8% (136/139) vs. 98.9% (375/379), respectively. Most isolates carried pfmdr1 wild-type at codon 86, with a significant difference in proportions genotypes (number of wild type/total sample) in samples collected during period 1 [92.9% (130/140)] compared with period 2 [96.9% (379/391)]. Investigation of pfmdr1 copy number was performed by real-time PCR. The proportions of isolates carried 1, 2, 3 and 4 or more than 4 copies of pfmdr1 (number of isolates carried correspondent copy number/total isolate) were significantly different between the two sample collecting periods (65.7% (90/137) vs. 87.8% (390/444), 18.2% (25/137) vs. 6.3%(28/444), 5.1% (7/137) vs. 1.4% (6/444) and 11.0% (15/137) vs. 4.5% (20/444), for period 1 vs. period 2, respectively). No pfk13 mutation was detected by nested PCR and nucleotide sequencing in all samples with successful analysis (n = 68). CONCLUSIONS The persistence of pfcrt mutations and pfmdr1 wild-types at codon 86, along with gene amplification in P. falciparum, contributes to the continued resistance of chloroquine and mefloquine in P. falciparum isolates in the study area. Regular surveillance of antimalarial drug resistance in P. falciparum, incorporating relevant molecular markers and treatment efficacy assessments, should be conducted.
Collapse
Affiliation(s)
- Phunuch Muhamad
- Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Pathumthani, 12120, Thailand
| | - Papichaya Phompradit
- Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Wanna Chaijaroenkul
- Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand
- Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Kesara Na-Bangchang
- Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Pathumthani, 12120, Thailand.
- Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
- Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
| |
Collapse
|
3
|
Suphakhonchuwong N, Rungsihirunrat K, Kuesap J. Surveillance of drug resistance molecular markers in Plasmodium vivax before and after introduction of dihydroartemisinin and piperaquine in Thailand: 2009-2019. Parasitol Res 2023; 122:2871-2883. [PMID: 37725258 DOI: 10.1007/s00436-023-07977-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
Resistance to antimalarial drugs is a serious issue around the world. Widespread Plasmodium vivax and P. falciparum coinfections are commonly found in Thailand. Dihydroartemisinin and piperaquine (DHA-PPQ) have been used as first-line treatments for P. falciparum since 2015, and chloroquine (CQ) and primaquine (PQ) have remained first-line drugs for P. vivax for more than 60 years. Coinfections may lead parasites to evolve with regard to genetics under selective drug pressure. This study is aimed at investigating genes linked to antimalarial resistance in P. vivax before and after introduction of DHA-PPQ as a new drug regimen in Thailand. A total of 400 P. vivax isolates were collected from samples along the Thai-Myanmar and Thai-Malaysian borders before (2009-2015) and after (2016-2019) introduction of DHA-PPQ. Genomic DNA of P. vivax was obtained and subjected to analysis of five drug resistance-associated genes (Pvdhfr, Pvdhps, Pvmdr1, Pvcrt-o, and PvK12) by nested polymerase chain reaction (PCR), restriction fragment length polymorphism (RFLP), and nucleotide sequencing. A high prevalence of Pvdhfr was found in both endemic areas over the period. The quadruple (57I/58R/61M/117T) Pvdhfr haplotype was predominant in both periods in both endemic areas. Although the wild-type haplotype of Pvdhps was predominant in Thai-Malaysian isolates in both periods, a single mutant haplotype (383G) was dominant in Thai-Myanmar isolates during both periods. A low prevalence of the Pvmdr1 976F mutation was found in both periods among Thai-Myanmar isolates. A significant decrease in Pvmdr1 976F was identified in Thai-Malaysian isolates from the second period (p < 0.01). Only one nonsynonymous mutation of Pvcrt-o (193E) and one synonymous mutation of PvK12 (R584) were detected in four isolates (4.7%) and one isolate (0.5%) in the first period among Thai-Myanmar isolates, respectively. Thus, with limited clinical efficacy data, the low prevalence of drug-resistance markers may suggest that there is a low prevalence of P. vivax-resistant strains and that the current drug regimen for P. vivax is still effective for treating this P. vivax parasite population. Continued surveillance of antimalarial drug resistance markers and monitoring of clinical drug efficacy should be conducted for epidemiological and policy implications.
Collapse
Affiliation(s)
| | | | - Jiraporn Kuesap
- Faculty of Allied Health Sciences, Thammasat University, Pathumthani, 12120, Thailand.
| |
Collapse
|
4
|
Looareesuwan P, Krudsood S, Lawpoolsri S, Tangpukdee N, Matsee W, Nguitragool W, Wilairatana P. Gametocyte prevalence and risk factors of P. falciparum malaria patients admitted at the Hospital for Tropical Diseases, Thailand: a 20-year retrospective study. Malar J 2023; 22:321. [PMID: 37872594 PMCID: PMC10591378 DOI: 10.1186/s12936-023-04728-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/26/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND The incidence of malaria in Thailand has dramatically declined over the past two decades, and the goal is to eliminate malaria by 2025. Despite significant progress, one of the key challenges to malaria elimination are undetected gametocyte carriers. Human migration adds complexity to the malaria situation, as it not only sustains local transmission but also poses the risk of spreading drug-resistant parasites. Currently, no study has assessed the prevalence of gametocytes across multiple years in Plasmodium falciparum malaria patients in Thailand, and the risk factors for gametocyte carriage have not been fully explored. METHODS Medical records of all P. falciparum malaria patients admitted from January 1, 2001 to December 31, 2020 at the Hospital for Tropical Diseases, Thailand, were retrospectively examined and a total of 1962 records were included for analysis. Both P. falciparum parasites and gametocytes were diagnosed by microscopy. A regression model was used to evaluate predictors of gametocyte carriage. RESULTS The study demonstrated gametocyte prevalence in low malaria transmission areas. Nine risk factors for gametocyte carriage were identified: age between 15 and 24 years [adjusted odds ratio (aOR) = 1.96, 95% confidence interval (CI) 1.18-3.26], Karen ethnicity (aOR = 2.59, 95% CI 1.56-4.29), preadmission duration of fever > 7 days (aOR = 5.40, 95% CI 3.92-7.41), fever on admission (> 37.5 °C) (aOR = 0.61, 95% CI 0.48-0.77), haemoglobin ≤ 8 g/dL (aOR = 3.32, 95% CI 2.06-5.33), asexual parasite density > 5000-25,000/µL (aOR = 0.71, 95% CI 0.52-0.98), asexual parasite density > 25,000-100,000/µL (aOR = 0.74, 95% CI 0.53-1.03), asexual parasite density > 100,000/µL (aOR = 0.51, 95% CI 0.36-0.72), platelet count ≤ 100,000/µL (aOR = 0.65, 95% CI 0.50-0.85, clinical features of severe malaria (aOR = 2.33, 95% CI 1.76-3.10) and dry season (aOR = 1.41, 95% CI 1.10-1.80). An increasing incidence of imported transnational malaria cases was observed over the past two decades. CONCLUSIONS This is the first study to determine the prevalence of gametocytes among patients with symptomatic P. falciparum malaria, identify the risk factors for gametocyte carriage, and potential gametocyte carriers in Thailand. Blocking transmission is one of the key strategies for eliminating malaria in these areas. The results might provide important information for targeting gametocyte carriers and improving the allocation of resources for malaria control in Thailand. This study supports the already nationally recommended use of a single dose of primaquine in symptomatic P. falciparum malaria patients to clear gametocytes.
Collapse
Affiliation(s)
- Panita Looareesuwan
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
- Thai Travel Clinic, Hospital for Tropical Diseases, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Srivicha Krudsood
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
- Clinical Malaria Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| | - Saranath Lawpoolsri
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Noppadon Tangpukdee
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Wasin Matsee
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Wang Nguitragool
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
5
|
Tuszewska H, Szczepański J, Mandziuk S, Trotsko N. Thiazolidin-4-one-based derivatives - Efficient tools for designing antiprotozoal agents. A review of the last decade. Bioorg Chem 2023; 133:106398. [PMID: 36739686 DOI: 10.1016/j.bioorg.2023.106398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/25/2022] [Accepted: 01/27/2023] [Indexed: 01/31/2023]
Abstract
Thiazolidin-4-one derivatives have a wide range of therapeutic implementations and clinical significance for medicinal chemistry. This heterocyclic ring has been reported to possess a variety of biological activities, including antiprotozoal activities that have inspired scientists to integrate this scaffold with different pharmacophoric fragments to design novel and effective antiprotozoal compounds. There are reviews describing thiazolidin-4-ones small molecules as good candidates with a single type of antiprotozoal activity, but none of these show collected news associated with the antiprotozoal activity of thiazolidin-4-ones and their SAR analysis from the last decade. In this review we are focusing on the antitoxoplasmic, anti-trypanosomal, antimalarial, antileishmanial, and antiamoebic activity of these derivatives, we attempt to summarize and analyze the recent developments with regard to the antiprotozoal potential of 4-TZD covering the structure-activity relationship and main molecular targets. The importance of various structural modifications at C2, N3, and C5 of the thiazolidine-4-one core has also been discussed in this review. We hope that all information concluded in this review can be useful for other researchers in constructing new effective antiprotozoal agents.
Collapse
Affiliation(s)
- Helena Tuszewska
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 4a, Chodzki Str., 20-093 Lublin, Poland
| | - Jacek Szczepański
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 4a, Chodzki Str., 20-093 Lublin, Poland
| | - Sławomir Mandziuk
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, 8, Jaczewski Str., 20-090 Lublin, Poland
| | - Nazar Trotsko
- Chair and Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 4a, Chodzki Str., 20-093 Lublin, Poland.
| |
Collapse
|
6
|
Bridging the Gap in Malaria Parasite Resistance, Current Interventions, and the Way Forward from in Silico Perspective: A Review. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227915. [PMID: 36432016 PMCID: PMC9692793 DOI: 10.3390/molecules27227915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/07/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
The past decade has seen most antimalarial drugs lose their clinical potency stemming from parasite resistance. Despite immense efforts by researchers to mitigate this global scourge, a breakthrough is yet to be achieved, as most current malaria chemotherapies suffer the same fate. Though the etiology of parasite resistance is not well understood, the parasite's complex life has been implicated. A drug-combination therapy with artemisinin as the central drug, artemisinin-based combination therapy (ACT), is currently the preferred malaria chemotherapy in most endemic zones. The emerging concern of parasite resistance to artemisinin, however, has compromised this treatment paradigm. Membrane-bound Ca2+-transporting ATPase and endocytosis pathway protein, Kelch13, among others, are identified as drivers in plasmodium parasite resistance to artemisinin. To mitigate parasite resistance to current chemotherapy, computer-aided drug design (CADD) techniques have been employed in the discovery of novel drug targets and the development of small molecule inhibitors to provide an intriguing alternative for malaria treatment. The evolution of plasmepsins, a class of aspartyl acid proteases, has gained tremendous attention in drug discovery, especially the non-food vacuole. They are expressed at multi-stage of the parasite's life cycle and involve in hepatocytes' egress, invasion, and dissemination of the parasite within the human host, further highlighting their essentiality. In silico exploration of non-food vacuole plasmepsin, PMIX and PMX unearthed the dual enzymatic inhibitory mechanism of the WM382 and 49c, novel plasmepsin inhibitors presently spearheading the search for potent antimalarial. These inhibitors impose structural compactness on the protease, distorting the characteristic twist motion. Pharmacophore modeling and structure activity of these compounds led to the generation of hits with better affinity and inhibitory prowess towards PMIX and PMX. Despite these headways, the major obstacle in targeting PM is the structural homogeneity among its members and to human Cathepsin D. The incorporation of CADD techniques described in the study at early stages of drug discovery could help in selective inhibition to augment malaria chemotherapy.
Collapse
|
7
|
Genome-wide functional screening of drug-resistance genes in Plasmodium falciparum. Nat Commun 2022; 13:6163. [PMID: 36257944 PMCID: PMC9579134 DOI: 10.1038/s41467-022-33804-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
The global spread of drug resistance is a major obstacle to the treatment of Plasmodium falciparum malaria. The identification of drug-resistance genes is an essential step toward solving the problem of drug resistance. Here, we report functional screening as a new approach with which to identify drug-resistance genes in P. falciparum. Specifically, a high-coverage genomic library of a drug-resistant strain is directly generated in a drug-sensitive strain, and the resistance gene is then identified from this library using drug screening. In a pilot experiment using the strain Dd2, the known chloroquine-resistant gene pfcrt is identified using the developed approach, which proves our experimental concept. Furthermore, we identify multidrug-resistant transporter 7 (pfmdr7) as a novel candidate for a mefloquine-resistance gene from a field-isolated parasite; we suggest that its upregulation possibly confers the mefloquine resistance. These results show the usefulness of functional screening as means by which to identify drug-resistance genes.
Collapse
|
8
|
Nana RRD, Bayengue SSB, Mogtomo MLK, Ngane ARN, Singh V. Anti-folate quintuple mutations in Plasmodium falciparum asymptomatic infections in Yaoundé, Cameroon. Parasitol Int 2022; 92:102657. [PMID: 36038059 DOI: 10.1016/j.parint.2022.102657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/07/2022] [Accepted: 08/19/2022] [Indexed: 11/28/2022]
Abstract
A major challenge in the fight to effectively control malaria is the emergence of resistant parasite to drugs used in therapy as well as for chemoprevention. In this study, single nucleotide polymorphisms (SNPs) associated with Plasmodium falciparum resistance to sulfadoxine-pyrimethamine (SP), one of the partner drugs in artemisinin-based therapies (ACTs) were studied in asymptomatic P. falciparum isolates from Cameroon. Dried Blood spots were collected from children with asymptomatic malaria enrolled during a household survey. The P. falciparum dihydrofolate reductase (Pfdhfr), dihydropteroate synthase (Pfdhps) and Kelch 13 genes were amplified and point mutations in these gene sequences were analyzed by sequencing. Among a total of 234 samples collected, 51 showed parasitaemia after microscopic examination of which 47 were P. falciparum mono-infections. Molecular analysis revealed 97.3% of mutant alleles at codons 51I, 59R and 108 N in Pfdhfr gene. In Pfdhps gene the most common mutation was 437G (83.3%); followed by 436A (47.6%) and 436F (28.6%). The association of mutations in the two genes (dhfr + dhps) showed 11 different haplotypes including three sextuple mutants (IRNI + AGKGA, IRNI + AAKGS, IRNI + AGKAS) and one septuple mutant (IRNI + AGKGS). For K13 gene no SNPs were seen in the studied asymptomatic malaria samples. The findings revealed presence of SP-resistant alleles in asymptomatic infected individuals with presence of sextuples and septuple SNPs. This emphasizes that regular profiling of antimalarial drugs resistance markers in such population is essential for malaria control and elimination programmes.
Collapse
Affiliation(s)
- Rodrigue Roman Dongang Nana
- Institute of Medical Research and Medicinal Plants studies, PO Box 13033, Yaoundé, Cameroon; ICMR-National Institute of Malaria Research, Dwarka, Sector 8, New Delhi 110077, India
| | | | | | - Anne Rosalie Ngono Ngane
- Department of Biochemistry, Faculty of Science, University of Douala, PO Box 24157, Douala, Cameroon
| | - Vineeta Singh
- ICMR-National Institute of Malaria Research, Dwarka, Sector 8, New Delhi 110077, India.
| |
Collapse
|
9
|
Assessing the diagnostic performance of a novel RT-PCR fluorescence method for the detection of human plasmodium species. PLoS One 2022; 17:e0272094. [PMID: 35925877 PMCID: PMC9352105 DOI: 10.1371/journal.pone.0272094] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 07/12/2022] [Indexed: 11/19/2022] Open
Abstract
Background Malaria elimination effort is hampered not only by the lack of effective medication but also due to the lack of sensitive diagnostic tools to detect infections with low levels of parasitemia. Therefore, more sensitive and specific high-throughput molecular diagnostic approaches are needed for accurate malaria diagnosis. Methods In the present study, the performance of a novel single-tube MC004 real-time polymerase chain reaction (PCR) assay (MRC-Holland, Amsterdam, the Netherlands) was assessed for the detection of infection and discrimination of Plasmodium species. Blood samples (n = 150) were collected from malaria suspected patients at Adama malaria diagnosis and treatment centre, Adama, central Ethiopia. The positive predictive value (PPV), negative predictive value (NPV), analytical sensitivity and specificity of the assay were assessed against the conventional microscopic method. Results Plasmodium species were detected in 59 (39.3%) of the samples by microscopy and in 62 (41.3%) by the novel MC004 RT-PCR. Plasmodium vivax, Plasmodium falciparum and mixed infections with Plasmodium falciparum & Plasmodium vivax accounted for 47.5%, 40.6% and 11.9% respectively as detected by microscopy. The MC004 RT-PCR assay identified 59.7% and 40.3% of the samples positive for Plasmodium vivax and Plasmodium falciparum respectively. The sensitivity, specificity, PPV, and NPV of the MC004 RT-PCR assay were 95.8%, 97.8%, 92%, and 98.9%, respectively. No mixed infections were detected using the MC004 assay. Conclusion The MC004 RT-PCR assay is a useful tool for the early detection of malaria and identification of Plasmodium species with a high degree of sensitivity and specificity. Due to its high sensitivity, and simplicity (being a single-tube assay), the MC004 is suitable for use in clinical settings and epidemiological studies.
Collapse
|
10
|
Recent Progress in the Development of Indole-Based Compounds Active against Malaria, Trypanosomiasis and Leishmaniasis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27010319. [PMID: 35011552 PMCID: PMC8746838 DOI: 10.3390/molecules27010319] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/31/2021] [Accepted: 01/01/2022] [Indexed: 01/06/2023]
Abstract
Human protozoan diseases represent a serious health problem worldwide, affecting mainly people in social and economic vulnerability. These diseases have attracted little investment in drug discovery, which is reflected in the limited available therapeutic arsenal. Authorized drugs present problems such as low efficacy in some stages of the disease or toxicity, which result in undesirable side effects and treatment abandonment. Moreover, the emergence of drug-resistant parasite strains makes necessary an even greater effort to develop safe and effective antiparasitic agents. Among the chemotypes investigated for parasitic diseases, the indole nucleus has emerged as a privileged molecular scaffold for the generation of new drug candidates. In this review, the authors provide an overview of the indole-based compounds developed against important parasitic diseases, namely malaria, trypanosomiasis and leishmaniasis, by focusing on the design, optimization and synthesis of the most relevant synthetic indole scaffolds recently reported.
Collapse
|
11
|
Berzosa P, Molina de la Fuente I, Ta-Tang TH, González V, García L, Rodríguez-Galet A, Díaz-Regañón R, Galán R, Cerrada-Gálvez L, Ncogo P, Riloha M, Benito A. Temporal evolution of the resistance genotypes of Plasmodium falciparum in isolates from Equatorial Guinea during 20 years (1999 to 2019). Malar J 2021; 20:463. [PMID: 34906159 PMCID: PMC8670137 DOI: 10.1186/s12936-021-04000-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/01/2021] [Indexed: 11/18/2022] Open
Abstract
Background Malaria is one of the deadliest diseases in the world, particularly in Africa. As such, resistance to anti-malarial drugs is one of the most important problems in terms of global malaria control. This study assesses the evolution of the different resistance markers over time and the possible influence of interventions and treatment changes that have been made in Equatorial Guinea. Methods A total of 1223 biological samples obtained in the period 1999 to 2019 were included in the study. Screening for mutations in the pfdhfr, pfdhps, pfmdr1, and pfcrt genes was carried out by nested PCR and restriction-fragment length polymorphisms (RFLPs), and the study of pfk13 genes was carried out by nested PCR, followed by sequencing to determine the presence of mutations. Results The partially and fully resistant haplotypes (pfdhfr + pfdhps) were found to increase over time. Moreover, in 2019, the fully resistant haplotype was found to be increasing, although its super-resistant counterpart remains much less prevalent. A continued decline in pfmdr1 and pfcrt gene mutations over time was also found. The number of mutations detected in pfk13 has increased since 2008, when artemisinin-based combination therapy (ACT) were first introduced, with more mutations being observed in 2019, with two synonymous and five non-synonymous mutations being detected, although these are not related to resistance to ACT. In addition, the non-synonymous A578S mutation, which is the most frequent on the African continent, was detected in 2013, although not in the following years. Conclusions Withdrawal of the use of chloroquine (CQ) as a treatment in Equatorial Guinea has been shown to be effective over time, as wild-type parasite populations outnumber mutant populations. The upward trend observed in sulfadoxine-pyrimethamine (SP) resistance markers suggest its misuse, either alone or in combination with artesunate (AS) or amodiaquine (AQ), in some areas of the country, as was found in a previous study conducted by this group, which allows selective pressure from SP to continue. Single nucleotide polymorphisms (SNPs) 540E and 581G do not exceed the limit of 50 and 10%, respectively, thus meaning that SP is still effective as an intermittent preventive treatment (IPT) in this country. As for the pfk13 gene, no mutations have been detected in relation to resistance to ACT. However, in 2019 there is a greater accumulation of non-synonymous mutations compared to years prior to 2008. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Pedro Berzosa
- National Centre of Tropical Medicine-Institute of Health Carlos III, Madrid, Spain.
| | - Irene Molina de la Fuente
- Department of Biomedicine and Biotechnology, University of Alcalá and National Centre of Tropical Medicine-Institute of Health Carlos III, Madrid, Spain
| | - Thuy-Huong Ta-Tang
- National Centre of Tropical Medicine-Institute of Health Carlos III, Madrid, Spain
| | - Vicenta González
- National Centre of Tropical Medicine-Institute of Health Carlos III, Madrid, Spain
| | - Luz García
- National Centre of Tropical Medicine-Institute of Health Carlos III, Madrid, Spain
| | - Ana Rodríguez-Galet
- National Centre of Tropical Medicine-Institute of Health Carlos III, Madrid, Spain.,HIV Molecular Epidemiology Laboratory, Ramón y Cajal-IRyCIS Hospital, Madrid, Spain
| | - Ramón Díaz-Regañón
- National Centre of Tropical Medicine-Institute of Health Carlos III, Madrid, Spain
| | - Rosario Galán
- National Centre of Tropical Medicine-Institute of Health Carlos III, Madrid, Spain
| | - Laura Cerrada-Gálvez
- National Centre of Tropical Medicine-Institute of Health Carlos III, Madrid, Spain
| | - Policarpo Ncogo
- State Foundation, Health, Childhood and Social Welfare FSP, Madrid, Spain
| | - Matilde Riloha
- Ministry of Health and Social Welfare-Malaria National Programme of Equatorial Guinea, Malabo, Equatorial Guinea
| | - Agustin Benito
- National Centre of Tropical Medicine-Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
12
|
Fink G, Venkataramani AS, Zanolini A. Early life adversity, biological adaptation, and human capital: evidence from an interrupted malaria control program in Zambia. JOURNAL OF HEALTH ECONOMICS 2021; 80:102532. [PMID: 34600186 DOI: 10.1016/j.jhealeco.2021.102532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/25/2021] [Accepted: 09/06/2021] [Indexed: 06/13/2023]
Abstract
Growing evidence from evolutionary biology demonstrates how early life shocks trigger physiological changes designed to be adaptive in challenging environments. We examine the implications of one type of physiological adaptation - immunity formation - for human capital accumulation. Using variation in early life malaria risk generated by an interrupted disease control program in Zambia, we show that exposure to infectious diseases during the first two years of life can reduce the harmful effects of malaria exposure on cognitive development during the preschool years. These findings suggest a non-linear and trajectory-dependent relationship between early life adversity and human capital formation.
Collapse
Affiliation(s)
- Günther Fink
- Swiss Tropical and Public Health Institute and University of Basel, Basel, Switzerland.
| | | | | |
Collapse
|
13
|
Rashidzadeh H, Tabatabaei Rezaei SJ, Adyani SM, Abazari M, Rahamooz Haghighi S, Abdollahi H, Ramazani A. Recent advances in targeting malaria with nanotechnology-based drug carriers. Pharm Dev Technol 2021; 26:807-823. [PMID: 34190000 DOI: 10.1080/10837450.2021.1948568] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Malaria, as one of the most common human infectious diseases, remains the greatest global health concern, since approximately 3.5 billion people around the world, especially those in subtropical areas, are at the risk of being infected by malaria. Due to the emergence and spread of drug resistance to the current antimalarials, malaria-related mortality and incidence rates have recently increased. To overcome the aforementioned obstacles, nano-vehicles based on biodegradable, natural, and non-toxic polymers have been developed. Accordingly, these systems are considered as a potential drug vehicle, which due to their unique properties such as the excellent safety profile, good biocompatibility, tunable structure, diversity, and the presence of functional groups within the polymer structure, could facilitate covalent attachment of targeting moieties and antimalarials to the polymeric nano-vehicles. In this review, we highlighted some recent developments of liposomes as unique nanoscale drug delivery vehicles and several polymeric nanovehicles, including hydrogels, dendrimers, self-assembled micelles, and polymer-drug conjugates for the effective delivery of antimalarials.
Collapse
Affiliation(s)
- Hamid Rashidzadeh
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran.,Laboratory of Novel Drug Delivery Systems, Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Jamal Tabatabaei Rezaei
- Laboratory of Novel Drug Delivery Systems, Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan, Iran
| | - Seyed Masih Adyani
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Morteza Abazari
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Samaneh Rahamooz Haghighi
- Department of Plant Production and Genetics, Faculty of Agriculture, University of Zanjan, Zanjan, Iran
| | - Hossien Abdollahi
- Department of Polymer Engineering, Faculty of Engineering, Urmia University, Urmia, Iran
| | - Ali Ramazani
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
14
|
Multidrug-Resistant Plasmodium falciparum Parasites in the Central Highlands of Vietnam Jeopardize Malaria Control and Elimination Strategies. Antimicrob Agents Chemother 2021; 65:AAC.01639-20. [PMID: 33526483 DOI: 10.1128/aac.01639-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/10/2021] [Indexed: 11/20/2022] Open
Abstract
Plasmodium falciparum resistance to dihydroartemisinin-piperaquine has spread through the Greater Mekong Subregion to southwestern Vietnam. In 2018 to 2019, we collected 127 P. falciparum isolates from Dak Nong (36), Dak Lak (55), Gia Lai (13), and Kon Tum (23) provinces in Vietnam's Central Highlands and found parasites bearing the Pfkelch13 C580Y mutation and multiple plasmepsin 2/3 genes (mean prevalence, 17.9%; range, 4.3% to 27.8%), conferring resistance to dihydroartemisinin-piperaquine. This information is important for drug policy decisions in Vietnam.
Collapse
|
15
|
Chawla J, Oberstaller J, Adams JH. Targeting Gametocytes of the Malaria Parasite Plasmodium falciparum in a Functional Genomics Era: Next Steps. Pathogens 2021; 10:346. [PMID: 33809464 PMCID: PMC7999360 DOI: 10.3390/pathogens10030346] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 02/04/2023] Open
Abstract
Mosquito transmission of the deadly malaria parasite Plasmodium falciparum is mediated by mature sexual forms (gametocytes). Circulating in the vertebrate host, relatively few intraerythrocytic gametocytes are picked up during a bloodmeal to continue sexual development in the mosquito vector. Human-to-vector transmission thus represents an infection bottleneck in the parasite's life cycle for therapeutic interventions to prevent malaria. Even though recent progress has been made in the identification of genetic factors linked to gametocytogenesis, a plethora of genes essential for sexual-stage development are yet to be unraveled. In this review, we revisit P. falciparum transmission biology by discussing targetable features of gametocytes and provide a perspective on a forward-genetic approach for identification of novel transmission-blocking candidates in the future.
Collapse
Affiliation(s)
- Jyotsna Chawla
- Molecular Medicine, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, MDC 7, Tampa, FL 33612, USA;
| | - Jenna Oberstaller
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Suite 404, Tampa, FL 33612, USA;
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Suite 404, Tampa, FL 33612, USA;
| |
Collapse
|
16
|
Kayode AT, Akano K, Ajogbasile FV, Uwanibe JN, Oluniyi PE, Bankole BE, Eromon PJ, Sowunmi A, Folarin OA, Volkman SK, McInnis B, Sabeti P, Wirth DF, Happi CT. Polymorphisms in Plasmodium falciparum chloroquine resistance transporter (Pfcrt) and multidrug-resistant gene 1 (Pfmdr-1) in Nigerian children 10 years post-adoption of artemisinin-based combination treatments. Int J Parasitol 2021; 51:301-310. [PMID: 33359205 PMCID: PMC7940560 DOI: 10.1016/j.ijpara.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 09/08/2020] [Accepted: 10/03/2020] [Indexed: 12/13/2022]
Abstract
The emergence and spread of Plasmodium falciparum parasites resistant to artemisinin derivatives and their partners in southeastern Asia threatens malaria control and elimination efforts, and heightens the need for an alternative therapy. We have explored the distribution of P. falciparum chloroquine resistance transporter (Pfcrt) and multidrug-resistant gene 1 (Pfmdr-1) haplotypes 10 years following adoption of artemisinin-based combination therapies in a bid to investigate the possible re-emergence of Chloroquine-sensitive parasites in Nigeria, and investigated the effect of these P. falciparum haplotypes on treatment outcomes of patients treated with artemisinin-based combination therapies. A total of 271 children aged <5 years with uncomplicated falciparum malaria were included in this study. Polymorphisms on codons 72-76 of the Pfcrt gene and codon 86 and 184 of Pfmdr-1 were determined using the high resolution melting assay. Of 240 (88.6%) samples successfully genotyped with HRM for Pfcrt, wildtype C72M74N75K76 (42.9%) and mutant C72I74E75T76 (53.8%) were observed. Also, wildtype N86Y184 (62.9%) and mutant N86F184 (21.1%), Y86Y184 (6.4%), and Y86F184 (0.4%) haplotypes of Pfmdr-1 were observed. Measures of responsiveness to ACTs were similar in children infected with P. falciparum crt haplotypes (C72I74E75T76 and C72M74N75K76) and major mdr-1 haplotypes (N86Y184, N86F184 and Y86Y184). Despite a 10 year gap since the malaria treatment policy changed to ACTs, over 50% of the P. falciparum parasites investigated in this study harboured the Chloroquine-resistant C72I74E75T76 haplotype, however this did not compromise the efficacy of artemisinin-based combination therapies. Should complete artemisinin resistance emerge from or spread to Nigeria, chloroquine might not be a good alternative therapy.
Collapse
Affiliation(s)
- Adeyemi T Kayode
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Kazeem Akano
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Fehintola V Ajogbasile
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Jessica N Uwanibe
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Paul E Oluniyi
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Bolajoko E Bankole
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Philomena J Eromon
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria
| | - Akintunde Sowunmi
- Institute of Medical Research and Training, College of Medicine, University of Ibadan; Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan, Nigeria
| | - Onikepe A Folarin
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria
| | - Sarah K Volkman
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Pardis Sabeti
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Dyann F Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Christian T Happi
- African Centre of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Nigeria; Department of Biological Sciences, Redeemer's University, Ede, Nigeria; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
17
|
Mensah BA, Aydemir O, Myers-Hansen JL, Opoku M, Hathaway NJ, Marsh PW, Anto F, Bailey J, Abuaku B, Ghansah A. Antimalarial Drug Resistance Profiling of Plasmodium falciparum Infections in Ghana Using Molecular Inversion Probes and Next-Generation Sequencing. Antimicrob Agents Chemother 2020; 64:e01423-19. [PMID: 31932374 PMCID: PMC7179265 DOI: 10.1128/aac.01423-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/19/2019] [Indexed: 01/24/2023] Open
Abstract
A key drawback to monitoring the emergence and spread of antimalarial drug resistance in sub-Saharan Africa is early detection and containment. Next-generation sequencing methods offer the resolution, sensitivity, and scale required to fill this gap by surveilling for molecular markers of drug resistance. We performed targeted sequencing using molecular inversion probes to interrogate five Plasmodium falciparum genes (pfcrt, pfmdr1, pfdhps, pfdhfr, and pfk13) implicated in chloroquine, sulfadoxine-pyrimethamine (SP), and artemisinin resistance in two sites in Ghana. A total of 803 dried blood spots from children aged between 6 months and 14 years presenting with uncomplicated P. falciparum malaria at the Begoro District Hospital in Begoro and the Ewim Polyclinic in Cape Coast, Ghana, from 2014 to 2017 were prepared on filter paper. Thirteen years after the removal of drug pressure, chloroquine-sensitive parasite strains with pfcrt K76 have increased nearly to fixation in Begoro, in the forest area (prevalence = 95%), but at a lower rate in Cape Coast, in the coastal region (prevalence = 71%, Z = -3.5, P < 0.001). In addition, pfmdr1 184F-bearing parasites are under strong selection. The pfdhfr/pfdhps quadruple genotype ( IRNG K), associated with SP resistance, is near saturation. Our study identified at a 2 to 10% prevalence pfdhps 581G, which is a sulfadoxine resistance marker that correlates with the failure of SP prophylaxis in pregnancy and which has not been observed in Ghana. The differences in the reexpansion of chloroquine-sensitive strains observed at the two study sites, the stronger SP resistance, and the high prevalence of pfmdr1 184F should be further monitored to inform malaria control strategies in Ghana.
Collapse
Affiliation(s)
- Benedicta A Mensah
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- School of Public Health, University of Ghana, Accra, Ghana
| | - Ozkan Aydemir
- Program in Bioinformatics and Integrative Biology, University of Massachusetts, Worcester, Massachusetts, USA
| | - James L Myers-Hansen
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Millicent Opoku
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Nicholas J Hathaway
- Program in Bioinformatics and Integrative Biology, University of Massachusetts, Worcester, Massachusetts, USA
| | - Patrick W Marsh
- Program in Bioinformatics and Integrative Biology, University of Massachusetts, Worcester, Massachusetts, USA
| | - Francis Anto
- School of Public Health, University of Ghana, Accra, Ghana
| | - Jeffrey Bailey
- Program in Bioinformatics and Integrative Biology, University of Massachusetts, Worcester, Massachusetts, USA
- Division of Transfusion Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Benjamin Abuaku
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Anita Ghansah
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
18
|
Exploration of copy number variation in genes related to anti-malarial drug resistance in Plasmodium falciparum. Gene 2020; 736:144414. [PMID: 32006594 DOI: 10.1016/j.gene.2020.144414] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022]
Abstract
Development of drug resistance in P. falciparum is one of the major problems associated with malaria treatment. Parasite genetic factors such as single nucleotide polymorphisms (SNPs) and copy number variations (CNV) have shown their role in drug resistance. Most of the studies have focused on the role of SNPs and drug resistance in parasite. However, it has also been shown that CNV is associated with adaptation and drug resistance in parasite. Hence, exploration of copy number polymorphism in essential genes of P. falciparum and their role in anti-malarial resistance is important. This review provides the recent information related to genetic profile of CNV marker in plasmepsin and other genes associated with drugresistanceinP. falciparum. It may be suggested that CNVs in plasmepsin genes are the major driver of piperaquine resistance. Moreover, CNVs in pfcrt and pfmdr1genes appear to play important role in adaptation and hence survival of the parasite. It may be hypothesized that targeting of CNV formation in the parasite could be beneficial for breakdown of its adaption in response to drug pressure.
Collapse
|
19
|
Kumar R, Sharma R, Kumar I, Upadhyay P, Dhiman AK, Kumar R, Kumar R, Purohit R, Sahal D, Sharma U. Evaluation of Antiplasmodial Potential of C2 and C8 Modified Quinolines: in vitro and in silico Study. Med Chem 2019; 15:790-800. [PMID: 30324888 DOI: 10.2174/1573406414666181015144413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/02/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Malaria remains a common life-threatening infectious disease across the globe due to the development of resistance by Plasmodium parasite against most antimalarial drugs. The situation demands new and effective drug candidates against Plasmodium. OBJECTIVES The objective of this study is to design, synthesize and test novel quinoline based molecules against the malaria parasite. METHODS C2 and C8 modified quinoline analogs obtained via C-H bond functionalization approach were synthesized and evaluated for inhibition of growth of P. falciparum grown in human red blood cells using SYBR Green microtiter plate based screening. Computational molecular docking studies were carried out with top fourteen molecules using Autodoc software. RESULTS The biological evaluation results revealed good activity of quinoline-8-acrylate 3f (IC50 14.2 µM), and the 2-quinoline-α-hydroxypropionates 4b (IC50 6.5 µM), 4j (IC50 5.5 µM) and 4g (IC50 9.5 µM), against chloroquine sensitive Pf3D7 strain. Top fourteen molecules were screened also against chloroquine resistant Pf INDO strain and the observed resistant indices were found to lie between 1 and 7.58. Computational molecular docking studies indicated a unique mode of binding of these quinolines to Falcipain-2 and heme moiety, indicating these to be the probable targets of their antiplasmodial action. CONCLUSION An important finding of our work is the fact that unlike Chloroquine which shows a resistance Index of 15, the resistance indices for the most promising molecules studied by us were about one indicating equal potency against drug sensitive and resistant strains of the malaria parasite.
Collapse
Affiliation(s)
- Rakesh Kumar
- Natural Product Chemistry and Process Development Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176 061, India.,Academy of Scientific and Innovative Research, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh-176 061, India
| | - Ritika Sharma
- Natural Product Chemistry and Process Development Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176 061, India.,Academy of Scientific and Innovative Research, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh-176 061, India
| | - Inder Kumar
- Natural Product Chemistry and Process Development Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176 061, India.,Academy of Scientific and Innovative Research, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh-176 061, India
| | - Pooja Upadhyay
- Malaria Drug Discovery Laboratory, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Ankit Kumar Dhiman
- Natural Product Chemistry and Process Development Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176 061, India.,Academy of Scientific and Innovative Research, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh-176 061, India
| | - Rohit Kumar
- Natural Product Chemistry and Process Development Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176 061, India
| | - Rakesh Kumar
- Natural Product Chemistry and Process Development Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176 061, India.,Centre for Nano and Material Sciences, Jain University, Jain Global, India
| | - Rituraj Purohit
- Academy of Scientific and Innovative Research, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh-176 061, India.,Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176 061, India
| | - Dinkar Sahal
- Malaria Drug Discovery Laboratory, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Upendra Sharma
- Natural Product Chemistry and Process Development Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176 061, India.,Academy of Scientific and Innovative Research, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh-176 061, India
| |
Collapse
|
20
|
Chhibber-Goel J, Sharma A. Profiles of Kelch mutations in Plasmodium falciparum across South Asia and their implications for tracking drug resistance. Int J Parasitol Drugs Drug Resist 2019; 11:49-58. [PMID: 31606696 PMCID: PMC6796718 DOI: 10.1016/j.ijpddr.2019.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 11/16/2022]
Abstract
Artemisinin-based combination therapy (ACT) offers highly successful treatment of malaria. Emergence and spread of Plasmodium falciparum (Pf) parasites with decreased susceptibility to ACT in South-East Asia has caused concern worldwide. The current accepted criteria to assess artemisinin (ART) resistance relies upon data on treatment failure, delayed parasite clearance at day 3 (DPC3), parasite clearance half-life (PCHL) and in-vitro/ex-vivo ring stage survival assays (RSAs). Interestingly, some studies suggest that DPC3 does not provide a distinct separation between ART sensitive/resistant strains, and RSA differences may also be inconclusive. More recently, recrudescence of ART treated Pf, independent of the presence of Kelch 13 (K13) mutation (C580Y), has been reported in the monkey malaria model suggesting that genes other than K13 like coronin, dhps, dhfr, crt, mdr1 and plasmepsin1 may contribute towards ACT failure. Here we have collated the distribution of K13 mutants from Pf strains in South Asia. A total of fifty Pf-K13 mutations have been studied for ART resistance in South Asia of which nine have been validated while eleven are potentials for ART resistance. The remaining thirty K13 mutations have been reported from various locations in South Asia but lack corroborative clinical data on ART resistance/ACT failure. Of the fifty, fourteen K13 mutations have been identified in India including four novel mutations (S549Y, G625R, N657H, D702N). Structural mapping of these K13 mutations does not offer any coherent explanation for their contribution towards ART resistance as they are scattered in the K13 structure. Thus, K13 mutations likely provide only a partial synopsis, and we propose that all suspect cases of ACT failure be assessed by: 1) DPC3, 2) PCHL, 3) in-vitro/ex-vivo RSAs and 4) GWAS data in an effort to annotate the resistance status of the parasites. These efforts may help in surveillance and containment of ART resistance/ACT failure in South Asia.
Collapse
Affiliation(s)
- Jyoti Chhibber-Goel
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India.
| | - Amit Sharma
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India.
| |
Collapse
|
21
|
Chaniad P, Techarang T, Phuwajaroanpong A, Punsawad C. Antimalarial Activity and Toxicological Assessment of Betula alnoides Extract against Plasmodium berghei Infections in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:2324679. [PMID: 31814839 PMCID: PMC6877991 DOI: 10.1155/2019/2324679] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/31/2019] [Indexed: 11/17/2022]
Abstract
The resistance of malaria parasites to the current antimalarial drugs has led to the search for novel effective drugs. Betula alnoides has been traditionally used for the treatment of malaria, but the scientific evidence to substantiate this claim is still lacking. Therefore, the present study aimed at evaluating the antimalarial activity and toxicity of an aqueous stem extract of B. alnoides in a mouse model. The in vivo antimalarial activity of an aqueous stem extract of B. alnoides was determined by a 4-day suppressive test in mice infected with chloroquine-sensitive Plasmodium berghei ANKA. The B. alnoides extract was administered orally at different doses of 200, 400, and 600 mg/kg body weight. The levels of parasitaemia, survival time, body weight change, and food and water consumption of the mice were determined. The acute toxicity of the extract was assessed in the mice for 14 days after the administration of a single oral dose of 5000 mg/kg. An aqueous stem extract of B. alnoides exhibited a significant dose-dependent reduction of parasitaemia in P. berghei-infected mice at all dose levels compared to the reduction in the negative control. Extract doses of 200, 400, and 600 mg/kg body weight suppressed the levels of parasitaemia by 46.90, 58.39, and 71.26%, respectively. The extract also significantly prolonged the survival times of the P. berghei-infected mice compared to the survival times of the negative control mice. In addition, at all dose levels, the extract prevented body weight loss in P. berghei-infected mice. For the acute toxicity, there were no significant alterations in the biochemical parameters and in the histopathology. In conclusion, the aqueous stem extract of B. alnoides possesses antimalarial properties. A single oral dose of 5000 mg/kg body weight had no significant toxic effects on the function and structure of the kidneys and liver. These results support its use in traditional medicine for the treatment of malaria.
Collapse
Affiliation(s)
- Prapaporn Chaniad
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Tropical Medicine Research Unit, Research Institute for Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Tachpon Techarang
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | | | - Chuchard Punsawad
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Tropical Medicine Research Unit, Research Institute for Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
22
|
Tan QW, Mutwil M. Inferring biosynthetic and gene regulatory networks from Artemisia annua RNA sequencing data on a credit card-sized ARM computer. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1863:194429. [PMID: 31634636 DOI: 10.1016/j.bbagrm.2019.194429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 02/05/2023]
Abstract
Prediction of gene function and gene regulatory networks is one of the most active topics in bioinformatics. The accumulation of publicly available gene expression data for hundreds of plant species, together with advances in bioinformatical methods and affordable computing, sets ingenuity as one of the major bottlenecks in understanding gene function and regulation. Here, we show how a credit card-sized computer retailing for <50 USD can be used to rapidly predict gene function and infer regulatory networks from RNA sequencing data. To achieve this, we constructed a bioinformatical pipeline that downloads and allows quality-control of RNA sequencing data; and generates a gene co-expression network that can reveal enzymes and transcription factors participating and controlling a given biosynthetic pathway. We exemplify this by first identifying genes and transcription factors involved in the biosynthesis of secondary cell wall in the plant Artemisia annua, the main natural source of the anti-malarial drug artemisinin. Networks were then used to dissect the artemisinin biosynthesis pathway, which suggest potential transcription factors regulating artemisinin biosynthesis. We provide the source code of our pipeline (https://github.com/mutwil/LSTrAP-Lite) and envision that the ubiquity of affordable computing, availability of biological data and increased bioinformatical training of biologists will transform the field of bioinformatics. This article is part of a Special Issue entitled: Transcriptional Profiles and Regulatory Gene Networks edited by Dr. Dr. Federico Manuel Giorgi and Dr. Shaun Mahony.
Collapse
Affiliation(s)
- Qiao Wen Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Marek Mutwil
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
23
|
Tawe L, Menegon M, Ramatlho P, Muthoga CW, Mutukwa N, Vurayai M, Bothudile W, Motshoge T, L'Episcopia M, Mosweunyane T, Kasvosve I, Severini C, Paganotti GM. Molecular Surveillance of Plasmodium falciparum Drug Resistance Markers in Clinical Samples from Botswana. Am J Trop Med Hyg 2019; 99:1499-1503. [PMID: 30350774 DOI: 10.4269/ajtmh.18-0440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Drug-resistant Plasmodium falciparum is a major threat to global malaria control and elimination efforts. In Botswana, a southern African country approaching malaria elimination, P. falciparum molecular data are not available. Parasites were assessed through pollymerase chain reaction (PCR) for confirmation of positive rapid diagnostic tests, multiplicity of infection (MOI), and drug resistance markers among isolates from clinical uncomplicated malaria cases collected at health facilities. Of 211 dried blood spot samples selected for the study, 186 (88.2%) were PCR positive for P. falciparum. The mean MOI based on MSP1 genotyping was 2.3 and was not associated with age. A high prevalence of wild-type parasites for pfcrt and pfmdr1 was found, with a haplotype frequency (K76/N86) of 88.8% and 17.7% of the isolates having two copies of the pfmdr1 gene. For pfATPase6, all the parasites carried the wild-type S769 allele. Sequencing showed no evidence of non-synonymous mutations associated with reduced artemisinin derivative sensitivity in the P. falciparum k13 gene. In conclusion, we found that P. falciparum parasites in Botswana were mostly wild type for the drug resistance markers evaluated. Yet, there was a high rate of a molecular marker associated to reduced sensitivity to lumefantrine. Our results indicate the need for systematic drug efficacy surveillance to complement malaria elimination efforts.
Collapse
Affiliation(s)
- Leabaneng Tawe
- Sub-Saharan African Network for TB/HIV Research Excellence (SANTHE) at Botswana-Harvard Partnership, Gaborone, Botswana.,Botswana-University of Pennsylvania Partnership, Gaborone, Botswana.,Department of Medical Laboratory Sciences, University of Botswana, Gaborone, Botswana
| | - Michela Menegon
- Department of Infectious Diseases, Istituto Superiore di Sanita', Rome, Italy
| | - Pleasure Ramatlho
- Department of Biological Sciences, University of Botswana, Gaborone, Botswana
| | | | - Naledi Mutukwa
- Department of Pathology, University of Botswana, Gaborone, Botswana
| | - Moses Vurayai
- National Health Laboratory, Department of Microbiology, Gaborone, Botswana
| | - Wame Bothudile
- Department of Medical Laboratory Sciences, University of Botswana, Gaborone, Botswana
| | - Thato Motshoge
- Department of Biological Sciences, University of Botswana, Gaborone, Botswana
| | | | | | - Ishmael Kasvosve
- Department of Medical Laboratory Sciences, University of Botswana, Gaborone, Botswana
| | - Carlo Severini
- Department of Infectious Diseases, Istituto Superiore di Sanita', Rome, Italy
| | - Giacomo M Paganotti
- Department of Biomedical Sciences, University of Botswana, Gaborone, Botswana.,Botswana-University of Pennsylvania Partnership, Gaborone, Botswana.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Beteck RM, Seldon R, Jordaan A, Warner DF, Hoppe HC, Laming D, Khanye SD. New Quinolone-Based Thiosemicarbazones Showing Activity Against Plasmodium falciparum and Mycobacterium tuberculosis. Molecules 2019; 24:molecules24091740. [PMID: 31060249 PMCID: PMC6540015 DOI: 10.3390/molecules24091740] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 11/30/2022] Open
Abstract
Co-infection of malaria and tuberculosis, although not thoroughly investigated, has been noted. With the increasing prevalence of tuberculosis in the African region, wherein malaria is endemic, it is intuitive to suggest that the probability of co-infection with these diseases is likely to increase. To avoid the issue of drug-drug interactions when managing co-infections, it is imperative to investigate new molecules with dual activities against the causal agents of these diseases. To this effect, a small library of quinolone-thiosemicarbazones was synthesised and evaluated in vitro against Plasmodium falciparum and Mycobacterium tuberculosis, the causal agents of malaria and tuberculosis, respectively. The compounds were also evaluated against HeLa cells for overt cytotoxicity. Most compounds in this series exhibited activities against both organisms, with compound 10, emerging as the hit; with an MIC90 of 2 µM against H37Rv strain of M. tuberculosis and an IC50 of 1 µM against the 3D7 strain of P. falciparum. This study highlights quinolone-thiosemicarabazones as a class of compounds that can be exploited further in search of novel, safe agents with potent activities against both the causal agents of malaria and tuberculosis.
Collapse
Affiliation(s)
- Richard M Beteck
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Rhodes University, Grahamstown 6140, South Africa.
| | - Ronnett Seldon
- Drug Discovery and Development Centre (H3-D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa.
| | - Audrey Jordaan
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town, Observatory 7925, South Africa.
| | - Digby F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town, Observatory 7925, South Africa.
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa.
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory 7925, South Africa.
| | - Heinrich C Hoppe
- Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Grahamstown 6140, South Africa.
- Centre for Chemico- and Biomedicinal Research, Rhodes University, Grahamstown 6140, South Africa.
| | - Dustin Laming
- Centre for Chemico- and Biomedicinal Research, Rhodes University, Grahamstown 6140, South Africa.
| | - Setshaba D Khanye
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Rhodes University, Grahamstown 6140, South Africa.
- Centre for Chemico- and Biomedicinal Research, Rhodes University, Grahamstown 6140, South Africa.
| |
Collapse
|
25
|
Scarpelli PH, Tessarin‐Almeida G, Viçoso KL, Lima WR, Borges‐Pereira L, Meissner KA, Wrenger C, Rafaello A, Rizzuto R, Pozzan T, Garcia CRS. Melatonin activates FIS1, DYN1, and DYN2 Plasmodium falciparum related-genes for mitochondria fission: Mitoemerald-GFP as a tool to visualize mitochondria structure. J Pineal Res 2019; 66:e12484. [PMID: 29480948 PMCID: PMC6585791 DOI: 10.1111/jpi.12484] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 02/01/2018] [Indexed: 02/06/2023]
Abstract
Malaria causes millions of deaths worldwide and is considered a huge burden to underdeveloped countries. The number of cases with resistance to all antimalarials is continuously increasing, making the identification of novel drugs a very urgent necessity. A potentially very interesting target for novel therapeutic intervention is the parasite mitochondrion. In this work, we studied in Plasmodium falciparum 3 genes coding for proteins homologues of the mammalian FIS1 (Mitochondrial Fission Protein 1) and DRP1 (Dynamin Related Protein 1) involved in mitochondrial fission. We studied the expression of P. falciparum genes that show ample sequence and structural homologies with the mammalian counterparts, namely FIS1, DYN1, and DYN2. The encoded proteins are characterized by a distinct pattern of expression throughout the erythrocytic cycle of P. falciparum, and their mRNAs are modulated by treating the parasite with the host hormone melatonin. We have previously reported that the knockout of the Plasmodium gene that codes for protein kinase 7 is essential for melatonin sensing. We here show that PfPk7 knockout results in major alterations of mitochondrial fission genes expression when compared to wild-type parasites, and no change in fission proteins expression upon treatment with the host hormone. Finally, we have compared the morphological characteristics (using MitoTracker Red CMX Ros) and oxygen consumption properties of P. falciparum mitochondria in wild-type parasites and PfPk7 Knockout strains. A novel GFP construct targeted to the mitochondrial matrix to wild-type parasites was also developed to visualize P. falciparum mitochondria. We here show that, the functional characteristics of P. falciparum are profoundly altered in cells lacking protein kinase 7, suggesting that this enzyme plays a major role in the control of mitochondrial morphogenesis and maturation during the intra-erythrocyte cell cycle progression.
Collapse
Affiliation(s)
- Pedro H. Scarpelli
- Departamento de ParasitologiaInstituto de Ciências BiomédicasUniversidade de São PauloSão PauloBrazil
- Departamento de FisiologiaInstituto de BiociênciasUniversidade de São PauloSão PauloBrazil
| | | | - Kênia Lopes Viçoso
- Departamento de FisiologiaInstituto de BiociênciasUniversidade de São PauloSão PauloBrazil
| | - Wania Rezende Lima
- Instituto de Ciências Exatas e Naturais‐MedicinaUniversidade Federal de Mato Grosso‐Campus RondonópolisMato GrossoBrazil
| | - Lucas Borges‐Pereira
- Departamento de FisiologiaInstituto de BiociênciasUniversidade de São PauloSão PauloBrazil
| | - Kamila Anna Meissner
- Departamento de ParasitologiaInstituto de Ciências BiomédicasUniversidade de São PauloSão PauloBrazil
| | - Carsten Wrenger
- Departamento de ParasitologiaInstituto de Ciências BiomédicasUniversidade de São PauloSão PauloBrazil
| | - Anna Rafaello
- CNR Neurosciences InstituteUniversity of PadovaPadovaItaly
| | | | - Tullio Pozzan
- CNR Neurosciences InstituteUniversity of PadovaPadovaItaly
| | - Celia R. S. Garcia
- Departamento de FisiologiaInstituto de BiociênciasUniversidade de São PauloSão PauloBrazil
- Departamento de Fisiologia, Instituto de BiociênciasUniversidade de São PauloSão PauloBrazil
| |
Collapse
|
26
|
von Seidlein L, Peto TJ, Landier J, Nguyen TN, Tripura R, Phommasone K, Pongvongsa T, Lwin KM, Keereecharoen L, Kajeechiwa L, Thwin MM, Parker DM, Wiladphaingern J, Nosten S, Proux S, Corbel V, Tuong-Vy N, Phuc-Nhi TL, Son DH, Huong-Thu PN, Tuyen NTK, Tien NT, Dong LT, Hue DV, Quang HH, Nguon C, Davoeung C, Rekol H, Adhikari B, Henriques G, Phongmany P, Suangkanarat P, Jeeyapant A, Vihokhern B, van der Pluijm RW, Lubell Y, White LJ, Aguas R, Promnarate C, Sirithiranont P, Malleret B, Rénia L, Onsjö C, Chan XH, Chalk J, Miotto O, Patumrat K, Chotivanich K, Hanboonkunupakarn B, Jittmala P, Kaehler N, Cheah PY, Pell C, Dhorda M, Imwong M, Snounou G, Mukaka M, Peerawaranun P, Lee SJ, Simpson JA, Pukrittayakamee S, Singhasivanon P, Grobusch MP, Cobelens F, Smithuis F, Newton PN, Thwaites GE, Day NPJ, Mayxay M, Hien TT, Nosten FH, Dondorp AM, White NJ. The impact of targeted malaria elimination with mass drug administrations on falciparum malaria in Southeast Asia: A cluster randomised trial. PLoS Med 2019; 16:e1002745. [PMID: 30768615 PMCID: PMC6377128 DOI: 10.1371/journal.pmed.1002745] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/15/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The emergence and spread of multidrug-resistant Plasmodium falciparum in the Greater Mekong Subregion (GMS) threatens global malaria elimination efforts. Mass drug administration (MDA), the presumptive antimalarial treatment of an entire population to clear the subclinical parasite reservoir, is a strategy to accelerate malaria elimination. We report a cluster randomised trial to assess the effectiveness of dihydroartemisinin-piperaquine (DP) MDA in reducing falciparum malaria incidence and prevalence in 16 remote village populations in Myanmar, Vietnam, Cambodia, and the Lao People's Democratic Republic, where artemisinin resistance is prevalent. METHODS AND FINDINGS After establishing vector control and community-based case management and following intensive community engagement, we used restricted randomisation within village pairs to select 8 villages to receive early DP MDA and 8 villages as controls for 12 months, after which the control villages received deferred DP MDA. The MDA comprised 3 monthly rounds of 3 daily doses of DP and, except in Cambodia, a single low dose of primaquine. We conducted exhaustive cross-sectional surveys of the entire population of each village at quarterly intervals using ultrasensitive quantitative PCR to detect Plasmodium infections. The study was conducted between May 2013 and July 2017. The investigators randomised 16 villages that had a total of 8,445 residents at the start of the study. Of these 8,445 residents, 4,135 (49%) residents living in 8 villages, plus an additional 288 newcomers to the villages, were randomised to receive early MDA; 3,790 out of the 4,423 (86%) participated in at least 1 MDA round, and 2,520 out of the 4,423 (57%) participated in all 3 rounds. The primary outcome, P. falciparum prevalence by month 3 (M3), fell by 92% (from 5.1% [171/3,340] to 0.4% [12/2,828]) in early MDA villages and by 29% (from 7.2% [246/3,405] to 5.1% [155/3,057]) in control villages. Over the following 9 months, the P. falciparum prevalence increased to 3.3% (96/2,881) in early MDA villages and to 6.1% (128/2,101) in control villages (adjusted incidence rate ratio 0.41 [95% CI 0.20 to 0.84]; p = 0.015). Individual protection was proportional to the number of completed MDA rounds. Of 221 participants with subclinical P. falciparum infections who participated in MDA and could be followed up, 207 (94%) cleared their infections, including 9 of 10 with artemisinin- and piperaquine-resistant infections. The DP MDAs were well tolerated; 6 severe adverse events were detected during the follow-up period, but none was attributable to the intervention. CONCLUSIONS Added to community-based basic malaria control measures, 3 monthly rounds of DP MDA reduced the incidence and prevalence of falciparum malaria over a 1-year period in areas affected by artemisinin resistance. P. falciparum infections returned during the follow-up period as the remaining infections spread and malaria was reintroduced from surrounding areas. Limitations of this study include a relatively small sample of villages, heterogeneity between villages, and mobility of villagers that may have limited the impact of the intervention. These results suggest that, if used as part of a comprehensive, well-organised, and well-resourced elimination programme, DP MDA can be a useful additional tool to accelerate malaria elimination. TRIAL REGISTRATION ClinicalTrials.gov NCT01872702.
Collapse
Affiliation(s)
- Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Thomas J. Peto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jordi Landier
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Institut de Recherche pour le Développement, Aix–Marseille University, INSERM, SESSTIM, Marseille, France
| | - Thuy-Nhien Nguyen
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Koukeo Phommasone
- Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
- Amsterdam Institute for Global Health & Development, Amsterdam, The Netherlands
| | - Tiengkham Pongvongsa
- Savannakhet Provincial Health Department, Savannakhet Province, Lao People’s Democratic Republic
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Khin Maung Lwin
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Lilly Keereecharoen
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Ladda Kajeechiwa
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - May Myo Thwin
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Daniel M. Parker
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Department of Population Health and Disease Prevention, University of California, Irvine, Irvine, California, United States of America
| | - Jacher Wiladphaingern
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Suphak Nosten
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Stephane Proux
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Vincent Corbel
- Maladies Infectieuses et Vecteurs: Écologie, Génétique, Evolution et Contrôle, Institut de Recherche pour le Développement, Université Montpellier, Montpellier, France
| | - Nguyen Tuong-Vy
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Truong Le Phuc-Nhi
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Do Hung Son
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Pham Nguyen Huong-Thu
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Kim Tuyen
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Nguyen Thanh Tien
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Le Thanh Dong
- Institute of Malariology, Parasitology, and Entomology, Ho Chi Minh City, Vietnam
| | - Dao Van Hue
- Center for Malariology, Parasitology and Entomology, Ninh Thuan Province, Vietnam
| | - Huynh Hong Quang
- Institute of Malariology, Parasitology, and Entomology, Quy Nhon, Vietnam
| | - Chea Nguon
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | | | - Huy Rekol
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Bipin Adhikari
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Gisela Henriques
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Pathogen Molecular Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Panom Phongmany
- Savannakhet Provincial Health Department, Savannakhet Province, Lao People’s Democratic Republic
| | - Preyanan Suangkanarat
- Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
| | - Atthanee Jeeyapant
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Benchawan Vihokhern
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Rob W. van der Pluijm
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Yoel Lubell
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lisa J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ricardo Aguas
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Cholrawee Promnarate
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- WWARN Asia Regional Centre, Mahidol University, Bangkok, Thailand
| | - Pasathorn Sirithiranont
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Benoit Malleret
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Laurent Rénia
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Carl Onsjö
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Faculty of Medicine and Health Sciences, Linköping University, Linköping, Linköping, Sweden
| | - Xin Hui Chan
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jeremy Chalk
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Olivo Miotto
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Krittaya Patumrat
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kesinee Chotivanich
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Borimas Hanboonkunupakarn
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Podjanee Jittmala
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nils Kaehler
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Christopher Pell
- Amsterdam Institute for Global Health & Development, Amsterdam, The Netherlands
| | - Mehul Dhorda
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- WWARN Asia Regional Centre, Mahidol University, Bangkok, Thailand
| | - Mallika Imwong
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Georges Snounou
- CEA–Université Paris Sud 11–INSERM U1184, IDMIT, Direction de la Recherche Fondamentale, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, Fontenay-aux-Roses, France
| | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Pimnara Peerawaranun
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sue J. Lee
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Julie A. Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Sasithon Pukrittayakamee
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Royal Society of Thailand, Bangkok, Thailand
| | - Pratap Singhasivanon
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Martin P. Grobusch
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Frank Cobelens
- Amsterdam Institute for Global Health & Development, Amsterdam, The Netherlands
| | | | - Paul N. Newton
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
| | - Guy E. Thwaites
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programmes, Ho Chi Minh City, Vietnam
| | - Nicholas P. J. Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mayfong Mayxay
- Lao–Oxford–Mahosot Hospital–Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao People’s Democratic Republic
- Institute of Research and Education Development, University of Health Sciences, Vientiane, Lao People’s Democratic Republic
| | - Tran Tinh Hien
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Institut de Recherche pour le Développement, Aix–Marseille University, INSERM, SESSTIM, Marseille, France
| | - Francois H. Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Arjen M. Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicholas J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
Santos APDAD, Fialho SN, Medeiros DSSD, Garay AFG, Diaz JAR, Gómez MCV, Teles CBG, Calderon LDA. Antiprotozoal action of synthetic cinnamic acid analogs. Rev Soc Bras Med Trop 2018; 51:849-853. [PMID: 30517542 DOI: 10.1590/0037-8682-0499-2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 06/05/2018] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Leishmaniasis, Chagas disease, and malaria cause morbidity globally. The drugs currently used for treatment have limitations. Activity of cinnamic acid analogs against Leishmania spp., Trypanosoma cruzi, and Plasmodium falciparum was evaluated in the interest of identifying new antiprotozoal compounds. METHODS In vitro effects of analogs against L. braziliensis, L. infantum chagasi, T. cruzi, and P. falciparum, and hemolytic and cytotoxic activities on NCTC 929 were determined. RESULTS Three analogs showed leishmanicidal and tripanocidal activity. No antiplasmodial, hemolytic, or cytotoxic activity was observed. CONCLUSIONS Antiprotozoal activity of analogs against L. infantum braziliensis, L. infantum chagasi, and T. cruzi was demonstrated.
Collapse
Affiliation(s)
- Ana Paula de Azevedo Dos Santos
- Plataforma de Bioensaios em Malária e Leishmaniose, Fundação Oswaldo Cruz, Porto Velho, RO, Brasil.,Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, RO, Brasil.,Centro de Estudos de Biomoléculas Aplicadas à Saúde, Universidade Federal de Rondônia, Porto Velho, RO, Brasil
| | - Saara Nery Fialho
- Plataforma de Bioensaios em Malária e Leishmaniose, Fundação Oswaldo Cruz, Porto Velho, RO, Brasil.,Centro Universitário São Lucas, Porto Velho, RO, Brasil
| | - Daniel Sol Sol de Medeiros
- Plataforma de Bioensaios em Malária e Leishmaniose, Fundação Oswaldo Cruz, Porto Velho, RO, Brasil.,Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, RO, Brasil.,Centro de Estudos de Biomoléculas Aplicadas à Saúde, Universidade Federal de Rondônia, Porto Velho, RO, Brasil
| | - Ana Fidelina Gómez Garay
- Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, RO, Brasil.,Centro de Estudos de Biomoléculas Aplicadas à Saúde, Universidade Federal de Rondônia, Porto Velho, RO, Brasil.,Centro para el Desarrollo de Investigación Científica, Asunción, Paraguay
| | - Jorge Alfonso Ruiz Diaz
- Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, RO, Brasil.,Centro de Estudos de Biomoléculas Aplicadas à Saúde, Universidade Federal de Rondônia, Porto Velho, RO, Brasil.,Centro para el Desarrollo de Investigación Científica, Asunción, Paraguay
| | | | - Carolina Bioni Garcia Teles
- Plataforma de Bioensaios em Malária e Leishmaniose, Fundação Oswaldo Cruz, Porto Velho, RO, Brasil.,Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, RO, Brasil.,Centro Universitário São Lucas, Porto Velho, RO, Brasil.,Instituto Nacional de Epidemiologia na Amazônia Ocidental, Porto Velho, RO, Brasil
| | - Leonardo de Azevedo Calderon
- Plataforma de Bioensaios em Malária e Leishmaniose, Fundação Oswaldo Cruz, Porto Velho, RO, Brasil.,Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, RO, Brasil.,Centro de Estudos de Biomoléculas Aplicadas à Saúde, Universidade Federal de Rondônia, Porto Velho, RO, Brasil
| |
Collapse
|
28
|
Garrido-Cardenas JA, Manzano-Agugliaro F, González-Cerón L, Gil-Montoya F, Alcayde-Garcia A, Novas N, Mesa-Valle C. The Identification of Scientific Communities and Their Approach to Worldwide Malaria Research. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15122703. [PMID: 30513616 PMCID: PMC6313382 DOI: 10.3390/ijerph15122703] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022]
Abstract
It is essential to establish a pattern to detect the strengths and weaknesses of working groups publishing on malaria, to promote coordination to facilitate the eradication of the disease. Given the complexity of the scientific network of groups and institutions studying malaria, it is necessary to use a mathematical algorithm that allows us to know the real structure of research on the disease in the world. In this work, articles with the word “malaria” in the title or author keywords gathered from Elsevier Scopus database were analyzed. By means of specific software, graphs were created. The analysis of the data allowed established different scientific communities, among which two were very diverse: one formed by those groups concerned about the vector transmission and control, and another one focused on the drug resistance of the parasite. Basic, applied, and operational research to eradicate malaria is an ambitious goal of the international institutions and the scientific community. The combination of effort and the establishment of a worldwide-scientific network that allows an effective interconnection (exchange) of knowledge, infrastructure technology, collaborators, financial resources, and datasets will contribute more effectively to end the disease.
Collapse
Affiliation(s)
| | | | - Lilia González-Cerón
- Regional Center for Public Health Research, National Institute of Public Health, Tapachula 30700, Chiapas, Mexico.
| | | | | | - Nuria Novas
- Department of Engineering, University of Almeria, CeiA3. 04120 Almeria, Spain.
| | | |
Collapse
|
29
|
Mason DJ, Eastman RT, Lewis RPI, Stott IP, Guha R, Bender A. Using Machine Learning to Predict Synergistic Antimalarial Compound Combinations With Novel Structures. Front Pharmacol 2018; 9:1096. [PMID: 30333748 PMCID: PMC6176478 DOI: 10.3389/fphar.2018.01096] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/07/2018] [Indexed: 01/28/2023] Open
Abstract
The parasite Plasmodium falciparum is the most lethal species of Plasmodium to cause serious malaria infection in humans, and with resistance developing rapidly novel treatment modalities are currently being sought, one of which being combinations of existing compounds. The discovery of combinations of antimalarial drugs that act synergistically with one another is hence of great importance; however an exhaustive experimental screen of large drug space in a pairwise manner is not an option. In this study we apply our machine learning approach, Combination Synergy Estimation (CoSynE), which can predict novel synergistic drug interactions using only prior experimental combination screening data and knowledge of compound molecular structures, to a dataset of 1,540 antimalarial drug combinations in which 22.2% were synergistic. Cross validation of our model showed that synergistic CoSynE predictions are enriched 2.74 × compared to random selection when both compounds in a predicted combination are known from other combinations among the training data, 2.36 × when only one compound is known from the training data, and 1.5 × for entirely novel combinations. We prospectively validated our model by making predictions for 185 combinations of 23 entirely novel compounds. CoSynE predicted 20 combinations to be synergistic, which was experimentally validated for nine of them (45%), corresponding to an enrichment of 1.70 × compared to random selection from this prospective data set. Such enrichment corresponds to a 41% reduction in experimental effort. Interestingly, we found that pairwise screening of the compounds CoSynE individually predicted to be synergistic would result in an enrichment of 1.36 × compared to random selection, indicating that synergy among compound combinations is not a random event. The nine novel and correctly predicted synergistic compound combinations mainly (where sufficient bioactivity information is available) consist of efflux or transporter inhibitors (such as hydroxyzine), combined with compounds exhibiting antimalarial activity alone (such as sorafenib, apicidin, or dihydroergotamine). However, not all compound synergies could be rationalized easily in this way. Overall, this study highlights the potential for predictive modeling to expedite the discovery of novel drug combinations in fight against antimalarial resistance, while the underlying approach is also generally applicable.
Collapse
Affiliation(s)
- Daniel J Mason
- Department of Chemistry, Centre for Molecular Informatics, University of Cambridge, Cambridge, United Kingdom.,Healx Ltd., Cambridge, United Kingdom
| | - Richard T Eastman
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Richard P I Lewis
- Department of Chemistry, Centre for Molecular Informatics, University of Cambridge, Cambridge, United Kingdom
| | - Ian P Stott
- Unilever Research and Development, Wirral, United Kingdom
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Andreas Bender
- Department of Chemistry, Centre for Molecular Informatics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
30
|
Feged-Rivadeneira A, Ángel A, González-Casabianca F, Rivera C. Malaria intensity in Colombia by regions and populations. PLoS One 2018; 13:e0203673. [PMID: 30208075 PMCID: PMC6135511 DOI: 10.1371/journal.pone.0203673] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/26/2018] [Indexed: 12/15/2022] Open
Abstract
Determining the distribution of disease prevalence among heterogeneous populations at the national scale is fundamental for epidemiology and public health. Here, we use a combination of methods (spatial scan statistic, topological data analysis and epidemic profile) to study measurable differences in malaria intensity by regions and populations of Colombia. This study explores three main questions: What are the regions of Colombia where malaria is epidemic? What are the regions and populations in Colombia where malaria is endemic? What associations exist between epidemic outbreaks between regions in Colombia? Plasmodium falciparum is most prevalent in the Pacific Coast, some regions of the Amazon Basin, and some regions of the Magdalena Basin. Plasmodium vivax is the most prevalent parasite in Colombia, particularly in the Northern Amazon Basin, the Caribbean, and municipalities of Sucre, Antioquia and Cordoba. We find an acute peak of malarial infection at 25 years of age. Indigenous and Afrocolombian populations experience endemic malaria (with household transmission). We find that Plasmodium vivax decreased in the most important hotspots, often with moderate urbanization rate, and was re-introduced to locations with moderate but sustained deforestation. Infection by Plasmodium falciparum, on the other hand, steadily increased in incidence in locations where it was introduced in the 2009-2010 generalized epidemic. Our findings suggest that Colombia is entering an unstable transmission state, where rapid decreases in one location of the country are interconnected with rapid increases in other parts of the country.
Collapse
Affiliation(s)
- Alejandro Feged-Rivadeneira
- Department of Anthropology, Stanford University, Stanford, CA, United States of America
- Department of Urban Management and Design, Universidad del Rosario, Bogotá, Colombia
- * E-mail:
| | - Andrés Ángel
- Department of Mathematics, Universidad de los Andes, Bogotá, Colombia
- Department of Mathematics and Statistics, Universidad del Norte, Barranquilla, Colombia
| | | | - Camilo Rivera
- Walmartlabs, Sunnyvale, CA, United States of America
| |
Collapse
|
31
|
Iwagami M, Tangpukdee N, Wilairatana P, Krudsood S, Dao LD, Nakazawa S, Sinuon M, Socheat D, Yasuoka J, Jimba M, Watanabe H, Kobayashi J, Toma H, Vanisaveth V, Hongvanthong B, Brey PT, Kano S. Pfcrt genotypes and related microsatellite DNA polymorphisms on Plasmodium falciparum differed among populations in the Greater Mekong Subregion. Parasitol Int 2018; 67:816-823. [PMID: 30165262 DOI: 10.1016/j.parint.2018.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 05/16/2018] [Accepted: 08/26/2018] [Indexed: 10/28/2022]
Abstract
Malaria morbidity and mortality have decreased gradually in the Greater Mekong Subregion (GMS). Presently, WHO sets a goal to eliminate malaria by 2030 in the GMS. However, drug-resistant malaria has been reported from several endemic areas. To achieve the goal of elimination, the status of the emergence and spread of drug resistance should be monitored. In this study, the genotype of the Plasmodium falciparum chloroquine (CQ) resistance transporter gene (pfcrt) and 6 microsatellite DNA loci flanking the gene were examined. P. falciparum isolates (n = 136) was collected from malaria patients in Thailand (n = 50, 2002-2005), Vietnam (n = 39, 2004), Laos (n = 15, 2007) and Cambodia (n = 32, 2009). Amino acid sequences at codons 72-76 on the gene were determined. All of the isolates from Thailand were CQ-resistant (CVIET), as were all of the isolates from Cambodia (CVIET, CVIDT). Thirteen of the 15 isolates (87%) from Laos were CQ-resistant (CVIET, CVIDT), whereas the other 2 (13%) were CQ-susceptible (CVMNK). In contrast, 27 of the 39 isolates (69%) from Vietnam were CQ-susceptible (CVMNK), whereas the other 12 (31%) were CQ-resistant (CVIET, CVIDT, CVMDT) or mixed (CVMNK/CVIDT). The mean of expected heterozygosity of the microsatellite loci was 0.444 in the Thai population, 0.482 in the Cambodian population, and 0.734 in the Vietnamese population. Genetic diversity in the Thai population was significantly lower than that in the Vietnamese population. These results suggested that chloroquine selective pressure on P. falciparum populations is heterogeneous in the GMS. Therefore, further examination to understand the mechanisms behind the emergence and spread of drug-resistant malaria are needed.
Collapse
Affiliation(s)
- Moritoshi Iwagami
- Department of Tropical Medicine and Malaria, National Center for Global Health and Medicine, Research Institute, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan; Ministry of Health, Institut Pasteur du Laos, Vientiane, Laos; SATREPS Project for Parasitic Diseases, Vientiane, Laos
| | - Noppadon Tangpukdee
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Srivicha Krudsood
- Department of Tropical Hygiene, Mahidol University, Bangkok, Thailand
| | - Le Duc Dao
- Molecular Biology Department, Parasitology and Entomology, National Institute of Malariology, Hanoi, Viet Nam
| | - Shusuke Nakazawa
- Department of Protozoology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Muth Sinuon
- National Centre for Parasitology, Entomology and Malaria Control, Ministry of Health, Phnom Penh, Cambodia
| | - Duong Socheat
- National Centre for Parasitology, Entomology and Malaria Control, Ministry of Health, Phnom Penh, Cambodia
| | - Junko Yasuoka
- Department of Community and Global Health, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masamine Jimba
- SATREPS Project for Parasitic Diseases, Vientiane, Laos; Department of Community and Global Health, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hisami Watanabe
- Division of Cellular and Molecular Immunology, Center of Molecular Biosciences, University of the Ryukyus, 207 Uehara, Nishihara-cho, Okinawa 903-0215, Japan
| | - Jun Kobayashi
- Department of Global Health, School of Health Sciences, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan
| | - Hiromu Toma
- Department of Parasitology and Immunopathoetiology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara-cho, Okinawa 903-0215, Japan
| | - Viengxay Vanisaveth
- Center of Malariology, Parasitology and Entomology, Ministry of Health, Vientiane, Laos
| | - Bouasy Hongvanthong
- SATREPS Project for Parasitic Diseases, Vientiane, Laos; Center of Malariology, Parasitology and Entomology, Ministry of Health, Vientiane, Laos
| | - Paul T Brey
- Ministry of Health, Institut Pasteur du Laos, Vientiane, Laos; SATREPS Project for Parasitic Diseases, Vientiane, Laos
| | - Shigeyuki Kano
- Department of Tropical Medicine and Malaria, National Center for Global Health and Medicine, Research Institute, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan; Ministry of Health, Institut Pasteur du Laos, Vientiane, Laos; SATREPS Project for Parasitic Diseases, Vientiane, Laos.
| |
Collapse
|
32
|
Krishnan KM, Williamson KC. The proteasome as a target to combat malaria: hits and misses. Transl Res 2018; 198:40-47. [PMID: 30009761 PMCID: PMC6422032 DOI: 10.1016/j.trsl.2018.04.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/26/2018] [Accepted: 04/30/2018] [Indexed: 01/25/2023]
Abstract
The proteasome plays a vital role throughout the life cycle as Plasmodium parasites quickly adapt to a new host and undergo a series of morphologic changes during asexual replication and sexual differentiation. Plasmodium carries 3 different types of protease complexes: typical eukaryotic proteasome (26S) that resides in the cytoplasm and the nucleus, a prokaryotic proteasome homolog ClpQ that resides in the mitochondria, and a caseinolytic protease complex ClpP that resides in the apicoplast. In silico prediction in conjunction with immunoprecipitation analysis of ubiquitin conjugates have suggested that over half of the Plasmodium falciparum proteome during asexual reproduction are potential targets for ubiquitination. The marked potency of multiple classes of proteasome inhibitors against all stages of the life cycle, synergy with the current frontline antimalarial, artemisinin, and recent advances identifying differences between Plasmodium and human proteasomes strongly support further drug development efforts.
Collapse
Affiliation(s)
| | - Kim C Williamson
- Uniformed Services University of the Health Sciences, Bethesda, Maryland.
| |
Collapse
|
33
|
Rogerio KR, Carvalho LJM, Domingues LHP, Neves BJ, Moreira Filho JT, Castro RN, Bianco Júnior C, Daniel-Ribeiro CT, Andrade CH, Graebin CS. Synthesis and molecular modelling studies of pyrimidinones and pyrrolo[3,4-d]-pyrimidinodiones as new antiplasmodial compounds. Mem Inst Oswaldo Cruz 2018; 113:e170452. [PMID: 29924131 PMCID: PMC6001580 DOI: 10.1590/0074-02760170452] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 05/10/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Malaria is responsible for 429,000 deaths per year worldwide, and more than 200 million cases were reported in 2015. Increasing parasite resistance has imposed restrictions to the currently available antimalarial drugs. Thus, the search for new, effective and safe antimalarial drugs is crucial. Heterocyclic compounds, such as dihydropyrimidinones (DHPM), synthesised via the Biginelli multicomponent reaction, as well as bicyclic compounds synthesised from DHPMs, have emerged as potential antimalarial candidates in the last few years. METHODS Thirty compounds were synthesised employing the Biginelli multicomponent reaction and subsequent one-pot substitution/cyclisation protocol; the compounds were then evaluated in vitro against chloroquine-resistant Plasmodium falciparum parasites (W2 strain). Drug cytotoxicity in baseline kidney African Green Monkey cells (BGM) was also evaluated. The most active in vitro compounds were evaluated against P. berghei parasites in mice. Additionally, we performed an in silico target fishing approach with the most active compounds, aiming to shed some light into the mechanism at a molecular level. RESULTS The synthetic route chosen was effective, leading to products with high purity and yields ranging from 10-84%. Three out of the 30 compounds tested were identified as active against the parasite and presented low toxicity. The in silico study suggested that among all the molecular targets identified by our target fishing approach, Protein Kinase 3 (PK5) and Glycogen Synthase Kinase 3β (GSK-3β) are the most likely molecular targets for the synthesised compounds. CONCLUSIONS We were able to easily obtain a collection of heterocyclic compounds with in vitro anti-P. falciparum activity that can be used as scaffolds for the design and development of new antiplasmodial drugs.
Collapse
Affiliation(s)
- Kamilla Rodrigues Rogerio
- Laboratório de Diversidade Molecular e Química Medicinal, Departamento de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brasil
| | - Leonardo J M Carvalho
- Laboratório de Pesquisas em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz-Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Luiza Helena Pinto Domingues
- Laboratório de Diversidade Molecular e Química Medicinal, Departamento de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brasil
| | - Bruno Junior Neves
- Laboratório de Planejamento de Fármacos e Modelagem Molecular, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brasil
| | - José Teófilo Moreira Filho
- Laboratório de Planejamento de Fármacos e Modelagem Molecular, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brasil
| | - Rosane Nora Castro
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brasil
| | - Cesare Bianco Júnior
- Laboratório de Pesquisas em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz-Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Claudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisas em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz-Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Carolina Horta Andrade
- Laboratório de Planejamento de Fármacos e Modelagem Molecular, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brasil
| | - Cedric Stephan Graebin
- Laboratório de Diversidade Molecular e Química Medicinal, Departamento de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brasil
| |
Collapse
|
34
|
Tantiamornkul K, Pumpaibool T, Piriyapongsa J, Culleton R, Lek-Uthai U. The prevalence of molecular markers of drug resistance in Plasmodium vivax from the border regions of Thailand in 2008 and 2014. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2018; 8:229-237. [PMID: 29677637 PMCID: PMC6039358 DOI: 10.1016/j.ijpddr.2018.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 02/08/2023]
Abstract
The prevalence of Plasmodium vivax is increasing in the border regions of Thailand; one potential problem confounding the control of malaria in these regions is the emergence and spread of drug resistance. The aim of this study was to determine the genetic diversity in genes potentially linked to drug resistance in P. vivax parasites isolated from four different border regions of Thailand; Thai-Myanmar (Tak, Mae Hong Son and Prachuap Khiri Khan Provinces), and Thai-Cambodian borders (Chanthaburi Province). Isolates were collected from 345 P. vivax patients in 2008 and 2014, and parasite DNA extracted and subjected to nucleotide sequencing at five putative drug-resistance loci (Pvdhfr, Pvdhps, Pvmdr1, Pvcrt-o and Pvk12). The prevalence of mutations in Pvdhfr, Pvdhps and Pvmdr1 were markedly different between the Thai-Myanmar and Thai-Cambodian border areas and also varied between sampling times. All isolates carried the Pvdhfr (58R and 117N/T) mutation, however, whereas the quadruple mutant allele (I57R58M61T117) was the most prevalent (69.6%) in the Thai-Myanmar border region, the double mutant allele (F57R58T61N117) was at fixation on the Thai-Cambodian border (100%). The most prevalent genotypes of Pvdhps and Pvmdr1 were the double mutant (S382G383K512G553) (65.1%) and single mutant (M958Y976F1076) (46.5%) alleles, respectively on the Thai-Myanmar border while the single Pvdhps mutant (S382G383K512A553) (52.7%) and the triple Pvmdr1 mutant (M958F976L1076) (81%) alleles were dominant on the Thai-Cambodian border. No mutations were observed in the Pvcrt-o gene in either region. Novel mutations in the Pvk12 gene, the P. vivax orthologue of PfK13, linked to artemisinin resistance in Plasmodium falciparum, were observed with three nonsynonymous and three synonymous mutations in six isolates (3.3%).
Collapse
Affiliation(s)
- Kritpaphat Tantiamornkul
- Department of Parasitology and Entomology, Faculty of Public Health, Mahidol University, Rajvithi Rd, Rajthewee District, Bangkok 10400, Thailand; Faculty of Graduate Studies, Mahidol University, Phuttamonthon 4 Rd, Nakorn Pathom 73170, Thailand
| | - Tepanata Pumpaibool
- College of Public Health Science, Chulalongkorn University, Phyathai Rd, Bangkok 10330, Thailand
| | - Jittima Piriyapongsa
- Genome Technology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani 12120, Thailand
| | - Richard Culleton
- Malaria Unit, Department of Pathology, Institute of Tropical Medicine, Nagasaki University, Sakamoto, Nagasaki 8528523, Japan.
| | - Usa Lek-Uthai
- Department of Parasitology and Entomology, Faculty of Public Health, Mahidol University, Rajvithi Rd, Rajthewee District, Bangkok 10400, Thailand.
| |
Collapse
|
35
|
De Lucia S, Tsamesidis I, Pau MC, Kesely KR, Pantaleo A, Turrini F. Induction of high tolerance to artemisinin by sub-lethal administration: A new in vitro model of P. falciparum. PLoS One 2018; 13:e0191084. [PMID: 29342187 PMCID: PMC5771598 DOI: 10.1371/journal.pone.0191084] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 12/18/2017] [Indexed: 12/13/2022] Open
Abstract
Artemisinin resistance is a major threat to malaria control efforts. Resistance is characterized by an increase in the Plasmodium falciparum parasite clearance half-life following treatment with artemisinin-based combination therapies (ACTs) and an increase in the percentage of surviving parasites. The remarkably short blood half-life of artemisinin derivatives may contribute to drug-resistance, possibly through factors including sub-lethal plasma concentrations and inadequate exposure. Here we selected for a new strain of artemisinin resistant parasites, termed the artemisinin resistant strain 1 (ARS1), by treating P. falciparum Palo Alto (PA) cultures with sub-lethal concentrations of dihydroartemisinin (DHA). The resistance phenotype was maintained for over 1 year through monthly maintenance treatments with low doses of 2.5 nM DHA. There was a moderate increase in the DHA IC50 in ARS1 when compared with parental strain PA after 72 h of drug exposure (from 0.68 nM to 2 nM DHA). In addition, ARS1 survived treatment physiologically relevant DHA concentrations (700 nM) observed in patients. Furthermore, we confirmed a lack of cross-resistance against a panel of antimalarials commonly used as partner drugs in ACTs. Finally, ARS1 did not contain Pfk13 propeller domain mutations associated with ART resistance in the Greater Mekong Region. With a stable growth rate, ARS1 represents a valuable tool for the development of new antimalarial compounds and studies to further elucidate the mechanisms of ART resistance.
Collapse
Affiliation(s)
- Serena De Lucia
- Department of Oncology, University of Turin, Turin, Italy
- * E-mail:
| | - Ioannis Tsamesidis
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Maria Carmina Pau
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Kristina R. Kesely
- Department of Biochemistry, Purdue University, West Lafayette, United States of America
| | - Antonella Pantaleo
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | |
Collapse
|
36
|
Dwivedi A, Reynes C, Kuehn A, Roche DB, Khim N, Hebrard M, Milanesi S, Rivals E, Frutos R, Menard D, Mamoun CB, Colinge J, Cornillot E. Functional analysis of Plasmodium falciparum subpopulations associated with artemisinin resistance in Cambodia. Malar J 2017; 16:493. [PMID: 29258508 PMCID: PMC5735551 DOI: 10.1186/s12936-017-2140-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 12/12/2017] [Indexed: 12/31/2022] Open
Abstract
Background Plasmodium falciparum malaria is one of the most widespread parasitic infections in humans and remains a leading global health concern. Malaria elimination efforts are threatened by the emergence and spread of resistance to artemisinin-based combination therapy, the first-line treatment of malaria. Promising molecular markers and pathways associated with artemisinin drug resistance have been identified, but the underlying molecular mechanisms of resistance remains unknown. The genomic data from early period of emergence of artemisinin resistance (2008–2011) was evaluated, with aim to define k13 associated genetic background in Cambodia, the country identified as epicentre of anti-malarial drug resistance, through characterization of 167 parasite isolates using a panel of 21,257 SNPs. Results Eight subpopulations were identified suggesting a process of acquisition of artemisinin resistance consistent with an emergence-selection-diffusion model, supported by the shifting balance theory. Identification of population specific mutations facilitated the characterization of a core set of 57 background genes associated with artemisinin resistance and associated pathways. The analysis indicates that the background of artemisinin resistance was not acquired after drug pressure, rather is the result of fixation followed by selection on the daughter subpopulations derived from the ancestral population. Conclusions Functional analysis of artemisinin resistance subpopulations illustrates the strong interplay between ubiquitination and cell division or differentiation in artemisinin resistant parasites. The relationship of these pathways with the P. falciparum resistant subpopulation and presence of drug resistance markers in addition to k13, highlights the major role of admixed parasite population in the diffusion of artemisinin resistant background. The diffusion of resistant genes in the Cambodian admixed population after selection resulted from mating of gametocytes of sensitive and resistant parasite populations. Electronic supplementary material The online version of this article (10.1186/s12936-017-2140-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ankit Dwivedi
- Institut de Biologie Computationnelle (IBC), 34095, Montpellier, France. .,Institut de Recherche en Cancérologie de Montpellier, Institut régional du Cancer Montpellier & Université de Montpellier, IRCM-INSERM U1194, 34298, Montpellier, France. .,Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Christelle Reynes
- Laboratoire de Biostatistiques, Informatique et Physique Pharmaceutique, UFR Pharmacie, Université de Montpellier, 34093, Montpellier, France.,Institut de Génomique Fonctionnelle-CNRS, 34094, Montpellier, France
| | - Axel Kuehn
- Institut de Recherche en Cancérologie de Montpellier, Institut régional du Cancer Montpellier & Université de Montpellier, IRCM-INSERM U1194, 34298, Montpellier, France
| | - Daniel B Roche
- Institut de Biologie Computationnelle (IBC), 34095, Montpellier, France.,Centre de Recherche en Biologie cellulaire de Montpellier, CNRS-UMR 5237, 34293, Montpellier, France
| | - Nimol Khim
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Maxim Hebrard
- Institut de Biologie Computationnelle (IBC), 34095, Montpellier, France.,Laboratoire d'informatique, de robotique et de microélectronique de Montpellier, LIRMM, CNRS, Université de Montpellier, 34095, Montpellier, France.,Center for Integrative Medical Sciences, RIKEN, Yokohama, Kanagawa, Japan
| | - Sylvain Milanesi
- Institut de Biologie Computationnelle (IBC), 34095, Montpellier, France
| | - Eric Rivals
- Institut de Biologie Computationnelle (IBC), 34095, Montpellier, France.,Laboratoire d'informatique, de robotique et de microélectronique de Montpellier, LIRMM, CNRS, Université de Montpellier, 34095, Montpellier, France
| | - Roger Frutos
- CIRAD, UMR Intertryp, 34398, Montpellier, France.,IES, UMR 5214, Université de Montpellier, CNRS, 34095, Montpellier, France
| | - Didier Menard
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia.,Biology of Host-Parasite Interactions Unit, Institut Pasteur, Paris, France
| | - Choukri Ben Mamoun
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Jacques Colinge
- Institut de Recherche en Cancérologie de Montpellier, Institut régional du Cancer Montpellier & Université de Montpellier, IRCM-INSERM U1194, 34298, Montpellier, France
| | - Emmanuel Cornillot
- Institut de Biologie Computationnelle (IBC), 34095, Montpellier, France. .,Institut de Recherche en Cancérologie de Montpellier, Institut régional du Cancer Montpellier & Université de Montpellier, IRCM-INSERM U1194, 34298, Montpellier, France.
| |
Collapse
|
37
|
Sweileh WM, Al-Jabi SW, Sawalha AF, AbuTaha AS, Zyoud SH. Bibliometric Analysis of Worldwide Publications on Antimalarial Drug Resistance (2006-2015). Malar Res Treat 2017; 2017:6429410. [PMID: 28856028 PMCID: PMC5569636 DOI: 10.1155/2017/6429410] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 04/28/2017] [Accepted: 07/10/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In response to international efforts to control and eradicate malaria, we designed this study to give a bibliometric overview of research productivity in antimalarial drug resistance (AMDR). METHODS Keywords related to AMDR were used to retrieve relevant literature using Scopus database. RESULTS A total of 976 publications with an h-index of 63 were retrieved. The number of publications showed a noticeable increase starting in the early 1990s. The USA was the most productive country with 337 publications equivalent to one-third of worldwide publications in this field. More than two-thirds of publications by the USA (236, 70.03%) were made by international collaboration. Of the top ten productive countries, two countries were from Mekong subregion, particularly Thailand and Cambodia. The Malaria Journal was the most productive journal (136, 13.93%) in this field. Mahidol University (80, 8.20%) in Thailand was the most productive institution. Seven articles in the top-ten list were about artemisinin resistance in Plasmodium falciparum, one was about chloroquine resistance, one was about sulfadoxine-pyrimethamine resistance, and the remaining one was about general multidrug resistance. CONCLUSION Eradication and control of AMDR require continuing research activity to help international health organizations identify spots that require an immediate action to implement appropriate measures.
Collapse
Affiliation(s)
- Waleed M. Sweileh
- Department of Physiology, Pharmacology and Toxicology, College of Medicine and Health Sciences, An-Najah National University, Nablus, State of Palestine
| | - Samah W. Al-Jabi
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, State of Palestine
| | - Ansam F. Sawalha
- Department of Physiology, Pharmacology and Toxicology, College of Medicine and Health Sciences, An-Najah National University, Nablus, State of Palestine
| | - Adham S. AbuTaha
- Department of Physiology, Pharmacology and Toxicology, College of Medicine and Health Sciences, An-Najah National University, Nablus, State of Palestine
| | - Sa'ed H. Zyoud
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, State of Palestine
| |
Collapse
|
38
|
Zhang J, Khan A, Kennard A, Grigg ME, Parkinson J. PopNet: A Markov Clustering Approach to Study Population Genetic Structure. Mol Biol Evol 2017; 34:1799-1811. [PMID: 28383661 PMCID: PMC5850731 DOI: 10.1093/molbev/msx110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
With the advent of low cost, high-throughput genome sequencing technology, population genomic data sets are being generated for hundreds of species of pathogenic, industrial, and agricultural importance. The challenge is how best to analyze and visually display these complex data sets to yield intuitive representations capable of capturing complex evolutionary relationships. Here we present PopNet, a novel computational method that identifies regions of shared ancestry in the chromosomes of related strains through clustering patterns of genetic variation. These relationships are subsequently visualized within a network by a novel implementation of chromosome painting. We apply PopNet to three diverse populations that feature differential rates of recombination and demonstrate its ability to capture evolutionary relationships as well as associate traits to specific loci. Compared with existing tools, PopNet provides substantial advances by both removing the need to predefine a single reference genome that can bias interpretation of population structure, as well as its ability to visualize multiple evolutionary relationships, such as recombination events and shared ancestry, across hundreds of strains.
Collapse
Affiliation(s)
- Javi Zhang
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Asis Khan
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD
| | - Andrea Kennard
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD
| | - Michael E. Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD
| | - John Parkinson
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ON, Canada
- Departments of Computer Science and Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
39
|
Tarique M, Chauhan M, Tuteja R. ATPase activity of Plasmodium falciparum MLH is inhibited by DNA-interacting ligands and dsRNAs of MLH along with UvrD curtail malaria parasite growth. PROTOPLASMA 2017; 254:1295-1305. [PMID: 27624787 DOI: 10.1007/s00709-016-1021-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 09/01/2016] [Indexed: 06/06/2023]
Abstract
Malaria caused by Plasmodium falciparum is the major disease burden all over the world. Recently, the situation has deteriorated because the malarial parasites are becoming progressively more resistant to numerous commonly used antimalarial drugs. Thus, there is a critical requirement to find other means to restrict and eliminate malaria. The mismatch repair (MMR) machinery of parasite is quite unique in several ways, and it can be exploited for finding new drug targets. MutL homolog (MLH) is one of the major components of MMR machinery, and along with UvrD, it helps in unwinding the DNA. We have screened several DNA-interacting ligands for their effect on intrinsic ATPase activity of PfMLH protein. This screening suggested that several ligands such as daunorubicin, etoposide, ethidium bromide, netropsin, and nogalamycin are inhibitors of the ATPase activity of PfMLH, and their apparent IC50 values range from 2.1 to 9.35 μM. In the presence of nogalamycin and netropsin, the effect was significant because in their presence, the V max value dropped from 1.024 μM of hydrolyzed ATP/min to 0.596 and 0.643 μM of hydrolyzed ATP/min, respectively. The effect of double-stranded RNAs of PfMLH and PfUvrD on growth of P. falciparum 3D7 strain was studied. The parasite growth was significantly inhibited suggesting that these components belonging to MMR pathway are crucial for the survival of the parasite.
Collapse
Affiliation(s)
- Mohammed Tarique
- Parasite Biology Group, International Centre for Genetic Engineering and Biotechnology, P. O. Box 10504, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Manish Chauhan
- Parasite Biology Group, International Centre for Genetic Engineering and Biotechnology, P. O. Box 10504, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Renu Tuteja
- Parasite Biology Group, International Centre for Genetic Engineering and Biotechnology, P. O. Box 10504, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| |
Collapse
|
40
|
O'Flaherty K, Maguire J, Simpson JA, Fowkes FJI. Immunity as a predictor of anti-malarial treatment failure: a systematic review. Malar J 2017; 16:158. [PMID: 28427418 PMCID: PMC5397737 DOI: 10.1186/s12936-017-1815-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 04/09/2017] [Indexed: 01/21/2023] Open
Abstract
Background Naturally acquired immunity can reduce parasitaemia and potentially influence anti-malarial treatment outcomes; however, evidence for this in the current literature provides conflicted results. The available evidence was synthesized to determine and quantify the association between host immunity and anti-malarial treatment failure. Methods Four databases were searched to identify studies investigating malaria antibody levels in patients receiving anti-malarial treatment for symptomatic malaria with treatment failure recorded according to the World Health Organization classification. Odds ratios or hazard ratios were extracted or calculated to quantify the association between malarial antibody levels and treatment failure, and findings from different studies were visualized using forest plots. Results Eight studies, including patients with falciparum malaria treated with mono- and combination therapy of artemisinin derivatives, sulfadoxine, pyrimethamine and chloroquine, were identified. Reported and calculated effect estimates varied greatly between studies, even those assessing the same antigens and treatments. An association between blood-stage IgG responses and treatment efficacy was observed. The greatest magnitudes of effect were observed for artemisinin [OR/HR (95% CI) range 0.02 (0.00, 0.45)–1.08 (0.57, 2.06)] and chloroquine [0.24 (0.04, 1.37)–0.32 (0.05, 1.96)] treatments, and larger magnitudes of effect were observed for variant surface antigen responses [0.02 (0.00, 0.45)–1.92 (0.94, 3.91)] when compared with merozoite specific responses [0.24 (0.04, 1.37)–2.83 (1.13, 7.09)]. Conclusions Naturally acquired malarial immunity is associated with reduced anti-malarial treatment failure in malaria endemic populations. Anti-malarial IgG effects treatment outcome differently for different anti-malarial drugs and antigen targets, and had the greatest impact during treatment with the current first-line treatments, the artemisinins. This has implications for the assessment of the therapeutic efficacy of anti-malarials, particularly in the context of emerging artemisinin resistance. Electronic supplementary material The online version of this article (doi:10.1186/s12936-017-1815-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katherine O'Flaherty
- Macfarlane Burnet Institute of Medical Research, Melbourne, VIC, 3004, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, 3010, Australia
| | - Julia Maguire
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, 3010, Australia
| | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, 3010, Australia
| | - Freya J I Fowkes
- Macfarlane Burnet Institute of Medical Research, Melbourne, VIC, 3004, Australia. .,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, 3010, Australia. .,Department of Epidemiology and Preventive Medicine and Department of Infectious Diseases, Monash University, Melbourne, 3800, Australia.
| |
Collapse
|
41
|
Reteng P, Vrisca V, Sukarno I, Djarkoni IH, Kalangi JA, Jacobs GE, Runtuwene LR, Eshita Y, Maeda R, Suzuki Y, Mongan AE, Warouw SM, Yamagishi J, Tuda J. Genetic polymorphisms in Plasmodium falciparum chloroquine resistance genes, pfcrt and pfmdr1, in North Sulawesi, Indonesia. BMC Res Notes 2017; 10:147. [PMID: 28376874 PMCID: PMC5379540 DOI: 10.1186/s13104-017-2468-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 03/24/2017] [Indexed: 01/25/2023] Open
Abstract
Background Malaria still poses one of the major threats to human health. Development of effective antimalarial drugs has decreased this threat; however, the emergence of drug-resistant Plasmodium falciparum, a cause of Malaria, is disconcerting. The antimalarial drug chloroquine has been effectively used, but resistant parasites have spread worldwide. Interestingly, the withdrawal of the drug reportedly leads to an increased population of susceptible parasites in some cases. We examined the prevalence of genomic polymorphisms in a malaria parasite P. falciparum, associated with resistance to an antimalarial drug chloroquine, after the withdrawal of the drug from Indonesia. Results Blood samples were collected from 95 malaria patients in North Sulawesi, Indonesia, in 2010. Parasite DNA was extracted and analyzed by polymerase chain reaction–restriction fragment length polymorphism (PCR–RFLP) for pfcrt and pfmdr1. In parallel, multiplex amplicon sequencing for the same genes was carried out with Illumina MiSeq. Of the 59 cases diagnosed as P. falciparum infection by microscopy, PCR–RFLP analysis clearly identified the genotype 76T in pfcrt in 44 cases. Sequencing analysis validated the identified genotypes in the 44 cases and demonstrated that the haplotype in the surrounding genomic region was exclusively SVMNT. Results of pfmdr1 were successfully obtained for 51 samples, where the genotyping results obtained by the two methods were completely consistent. In pfmdr1, the 86Y mutant genotype was observed in 45 cases (88.2%). Conclusions Our results suggest that the prevalence of the mutated genotypes remained dominant even 6 years after the withdrawal of chloroquine from this region. Diversified haplotype of the resistance-related locus, potentially involved in fitness costs, unauthorized usage of chloroquine, and/or a short post-withdrawal period may account for the observed high persistence of prevalence. Electronic supplementary material The online version of this article (doi:10.1186/s13104-017-2468-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Patrick Reteng
- Department of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu, Manado, 95115, Indonesia
| | - Visia Vrisca
- Department of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu, Manado, 95115, Indonesia
| | - Inka Sukarno
- Department of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu, Manado, 95115, Indonesia
| | - Ilham Habib Djarkoni
- Department of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu, Manado, 95115, Indonesia
| | - Jane Angela Kalangi
- Department of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu, Manado, 95115, Indonesia
| | - George Eduardo Jacobs
- Department of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu, Manado, 95115, Indonesia
| | - Lucky Ronald Runtuwene
- Department of Medical Genome Sciences, University of Tokyo, Kashiwa, Chiba, 277-8562, Japan
| | - Yuki Eshita
- Faculty of Medicine, Oita University, Yufu, Oita, 879-5593, Japan.,Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.,Research Center for Zoonosis Control, Hokkaido University, North 20, West 10 Kita-ku, Sapporo, Hokkaido, 001-0020, Japan
| | - Ryuichiro Maeda
- Department of Human Sciences, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, University of Tokyo, Kashiwa, Chiba, 277-8562, Japan
| | - Arthur Elia Mongan
- Department of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu, Manado, 95115, Indonesia
| | - Sarah Maria Warouw
- Department of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu, Manado, 95115, Indonesia
| | - Junya Yamagishi
- Research Center for Zoonosis Control, Hokkaido University, North 20, West 10 Kita-ku, Sapporo, Hokkaido, 001-0020, Japan. .,Global Station for Zoonosis Control, GI-CoRE, Hokkaido University, North 20, West 10 Kita-ku, Sapporo, Hokkaido, 001-0020, Japan.
| | - Josef Tuda
- Department of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu, Manado, 95115, Indonesia
| |
Collapse
|
42
|
Hakamada K, Watanabe H, Kawano R, Noguchi K, Yohda M. Expression and characterization of the Plasmodium translocon of the exported proteins component EXP2. Biochem Biophys Res Commun 2017; 482:700-705. [DOI: 10.1016/j.bbrc.2016.11.097] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 11/16/2016] [Indexed: 10/20/2022]
|
43
|
Risk of drug resistance in Plasmodium falciparum malaria therapy-a systematic review and meta-analysis. Parasitol Res 2016; 116:781-788. [PMID: 28028628 DOI: 10.1007/s00436-016-5353-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 12/13/2016] [Indexed: 12/30/2022]
Abstract
Plasmodium falciparum is responsible for the vast majority of the morbidity and mortality associated with malaria infection globally. Although a number of studies have reported the emergence of drug resistance in different therapies for P. falciparum infection, the degree of the drug resistance in different antimalarials is still unclear. This research investigated the risk of drug resistance in the therapies with different medications based on meta-analyses. Relevant original randomized control trials (RCTs) were searched in all available electronic databases. Pooled relative risks (RRs) with 95% confidence intervals (95% CIs) were used to evaluate the risk of drug resistance resulting from different treatments. Seventy-eight studies were included in the meta-analysis to compare drug resistance in the treatment of P. falciparum infections and yielded the following results: chloroquine (CQ) > sulfadoxine-pyrimethamine (SP) (RR = 3.67, p < 0.001 ), mefloquine (MQ) < SP (RR = 0.26, p < 0.001), artesunate + sulfadoxine-pyrimethamine (AS + SP) > artemether + lumefantrine (AL) (RR = 2.94, p < 0.001), dihydroartemisinin + piperaquine (DHA + PQ) < AL (RR = 0.7, p < 0.05), and non-artemisinin-based combination therapies (NACTs) > artemisinin-based combination therapies (ACTs) (RR = 1.93, p < 0.001); no significant difference was found in amodiaquine (AQ) vs. SP, AS + AQ vs. AS + SP, AS + AQ vs. AL, or AS + MQ vs. AL. These results presented a global view for the current status of antimalarial drug resistance and provided a guidance for choice of antimalarials for efficient treatment and prolonging the life span of the current effective antimalarial drugs.
Collapse
|
44
|
Magistrado PA, Corey VC, Lukens AK, LaMonte G, Sasaki E, Meister S, Wree M, Winzeler E, Wirth DF. Plasmodium falciparum Cyclic Amine Resistance Locus (PfCARL), a Resistance Mechanism for Two Distinct Compound Classes. ACS Infect Dis 2016; 2:816-826. [PMID: 27933786 PMCID: PMC5109296 DOI: 10.1021/acsinfecdis.6b00025] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
![]()
MMV007564
is a novel antimalarial benzimidazolyl piperidine chemotype
identified in cellular screens. To identify the genetic determinant
of MMV007564 resistance, parasites were cultured in the presence of
the compound to generate resistant lines. Whole genome sequencing
revealed distinct mutations in the gene named Plasmodium
falciparum cyclic amine resistance locus (pfcarl), encoding a conserved protein of unknown function.
Mutations in pfcarl are strongly associated with
resistance to a structurally unrelated class of compounds, the imidazolopiperazines,
including KAF156, currently in clinical trials. Our data demonstrate
that pfcarl mutations confer resistance to two distinct
compound classes, benzimidazolyl piperidines and imidazolopiperazines.
However, MMV007564 and the imidazolopiperazines, KAF156 and GNF179,
have different timings of action in the asexual blood stage and different
potencies against the liver and sexual blood stages. These data suggest
that pfcarl is a multidrug-resistance gene rather
than a common target for benzimidazolyl piperidines and imidazolopiperazines.
Collapse
Affiliation(s)
- Pamela A. Magistrado
- Department
of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| | - Victoria C. Corey
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Amanda K. Lukens
- Department
of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, United States
- Infectious
Disease Program, The Broad Institute, 415 Main Street, Cambridge, Massachusetts 02142, United States
| | - Greg LaMonte
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Erika Sasaki
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Stephan Meister
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Melanie Wree
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Elizabeth Winzeler
- School
of Medicine, University of California—San Diego, La Jolla, California 92093, United States
| | - Dyann F. Wirth
- Department
of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, United States
- Infectious
Disease Program, The Broad Institute, 415 Main Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
45
|
Kume A, Anh DTM, Shichiri M, Ishida N, Suzuki H. Probucol dramatically enhances dihydroartemisinin effect in murine malaria. Malar J 2016; 15:472. [PMID: 27634686 PMCID: PMC5025592 DOI: 10.1186/s12936-016-1532-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/09/2016] [Indexed: 12/22/2022] Open
Abstract
Background Artemisinin-based combination therapy (ACT) has been adopted as national policy for the first-line treatment in large number of malaria-endemic regions. However, artemisinin-resistant parasites have emerged and are spreading, with slow-cleaning parasites being reported in patients treated with ACT. It means that more parasites are exposed to the partner drug alone and the risk of developing resistant parasites against the partner drug is increasing. Therefore, the development of a new method to enhance the effect of artemisinin is required. In this study, the potential effect of probucol as a combination drug of dihydroartemisinin (DHA), an artemisinin derivative, was examined. Methods C57BL/6 J mice infected with Plasmodium yoelii XL-17 were treated with probucol and/or DHA. The mice were fed with a probucol mixed diet from 2 weeks before infection and through infection period. DHA was injected to mice three to 5 days post infection once a day. In addition, 0.5 % (w/w) probucol was mixed with vitamin E supplemented diet (800 mg/kg) and fed to mice infected with P. yoelii XL-17 to examine the mechanisms of probucol on murine malaria. Furthermore, 8-OHdG, a biomarker of oxidized DNA, was detected in infected red blood cells (iRBC) taken from infected mice by immunofluorescent staining. Results With dose-dependent manner, both probucol and DHA decreased parasitaemia and increased survival rate of mice infected with P. yoelii XL-17. A significantly larger amount of 8-OHdG was detected in iRBC taking from probucol-treated mice than control mice. In addition, a large amount of vitamin E supplementation eliminated the effect of probucol against P. yoelii XL-17 infection and lowered the effect of probucol on host plasma vitamin E concentration. The effective doses for probucol and DHA were 0.5 % and 30 mg/kg, respectively, in each single treatment. While the combination treatment of 0.25 % probucol and 7.5 mg/kg DHA was effective in all mice from P. yoelii XL-17 infection. Conclusion This study demonstrated that probucol has some impact on malaria by oxidative stress through the induction of host plasma vitamin E deficiency. Moreover, the effective dose of DHA on malaria was decreased by prophylactic treatment of probucol. This finding indicates that probucol might be a candidate for a prophylactic treatment drug to enhance the effect of DHA.
Collapse
Affiliation(s)
- Aiko Kume
- Research Unit for Functional Genomics, National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro City, Hokkaido, Japan.,The United Graduate School of Veterinary Sciences, Gifu University, Gifu City, Japan
| | - Dang Trinh Minh Anh
- Research Unit for Functional Genomics, National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro City, Hokkaido, Japan.,Microbiology and Immunology Department, Pasteur Institute in Ho Chi Minh City, Ministry of Health, Ho Chi Minh City, Vietnam
| | - Mototada Shichiri
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ikeda City, Osaka, Japan
| | - Noriko Ishida
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ikeda City, Osaka, Japan
| | - Hiroshi Suzuki
- Research Unit for Functional Genomics, National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro City, Hokkaido, Japan. .,The United Graduate School of Veterinary Sciences, Gifu University, Gifu City, Japan.
| |
Collapse
|
46
|
Alout H, Dabiré RK, Djogbénou LS, Abate L, Corbel V, Chandre F, Cohuet A. Interactive cost of Plasmodium infection and insecticide resistance in the malaria vector Anopheles gambiae. Sci Rep 2016; 6:29755. [PMID: 27432257 PMCID: PMC4949420 DOI: 10.1038/srep29755] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/23/2016] [Indexed: 01/19/2023] Open
Abstract
Insecticide resistance raises concerns for the control of vector-borne diseases. However, its impact on parasite transmission could be diverse when considering the ecological interactions between vector and parasite. Thus we investigated the fitness cost associated with insecticide resistance and Plasmodium falciparum infection as well as their interactive cost on Anopheles gambiae survival and fecundity. In absence of infection, we observed a cost on fecundity associated with insecticide resistance. However, survival was higher for mosquito bearing the kdr mutation and equal for those with the ace-1(R) mutation compared to their insecticide susceptible counterparts. Interestingly, Plasmodium infection reduced survival only in the insecticide resistant strains but not in the susceptible one and infection was associated with an increase in fecundity independently of the strain considered. This study provides evidence for a survival cost associated with infection by Plasmodium parasite only in mosquito selected for insecticide resistance. This suggests that the selection of insecticide resistance mutation may have disturbed the interaction between parasites and vectors, resulting in increased cost of infection. Considering the fitness cost as well as other ecological aspects of this natural mosquito-parasite combination is important to predict the epidemiological impact of insecticide resistance.
Collapse
Affiliation(s)
- Haoues Alout
- Institut de recherche pour le développement (IRD), Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), UM1-UM2-CNRS 5290 IRD 224, Montpellier, France
| | - Roch K. Dabiré
- Institut de Recherche en Sciences de la Santé (IRSS), 01 BP 545 Bobo-Dioulasso 01, Burkina Faso
| | - Luc S. Djogbénou
- Institut Régional de Santé Publique/Université d’Abomey-Calavi, 01 BP 918 Cotonou, Bénin
| | - Luc Abate
- Institut de recherche pour le développement (IRD), Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), UM1-UM2-CNRS 5290 IRD 224, Montpellier, France
| | - Vincent Corbel
- Institut de recherche pour le développement (IRD), Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), UM1-UM2-CNRS 5290 IRD 224, Montpellier, France
- Department of Entomology, Faculty of Agriculture, Kasetsart University, Bangkok 10900, Thailand
| | - Fabrice Chandre
- Institut de recherche pour le développement (IRD), Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), UM1-UM2-CNRS 5290 IRD 224, Montpellier, France
| | - Anna Cohuet
- Institut de recherche pour le développement (IRD), Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle (MIVEGEC), UM1-UM2-CNRS 5290 IRD 224, Montpellier, France
| |
Collapse
|
47
|
Shitlani D, Choudhary R, Pandey DP, Bodakhe SH. Ameliorative antimalarial effects of the combination of rutin and swertiamarin on malarial parasites. ASIAN PACIFIC JOURNAL OF TROPICAL DISEASE 2016. [DOI: 10.1016/s2222-1808(16)61067-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
48
|
Chang HH, Childs LM, Buckee CO. Variation in infection length and superinfection enhance selection efficiency in the human malaria parasite. Sci Rep 2016; 6:26370. [PMID: 27193195 PMCID: PMC4872237 DOI: 10.1038/srep26370] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/28/2016] [Indexed: 01/08/2023] Open
Abstract
The capacity for adaptation is central to the evolutionary success of the human malaria parasite Plasmodium falciparum. Malaria epidemiology is characterized by the circulation of multiple, genetically diverse parasite clones, frequent superinfection, and highly variable infection lengths, a large number of which are chronic and asymptomatic. The impact of these characteristics on the evolution of the parasite is largely unknown, however, hampering our understanding of the impact of interventions and the emergence of drug resistance. In particular, standard population genetic frameworks do not accommodate variation in infection length or superinfection. Here, we develop a population genetic model of malaria including these variations, and show that these aspects of malaria infection dynamics enhance both the probability and speed of fixation for beneficial alleles in complex and non-intuitive ways. We find that populations containing a mixture of short- and long-lived infections promote selection efficiency. Interestingly, this increase in selection efficiency occurs even when only a small fraction of the infections are chronic, suggesting that selection can occur efficiently in areas of low transmission intensity, providing a hypothesis for the repeated emergence of drug resistance in the low transmission setting of Southeast Asia.
Collapse
Affiliation(s)
- Hsiao-Han Chang
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Lauren M Childs
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Caroline O Buckee
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
49
|
Paczkowski M, Mwandama D, Marthey D, Luka M, Makuta G, Sande J, Ali D, Troell P, Mathanga DP, Gutman J. In vivo efficacy of artemether-lumefantrine and artesunate-amodiaquine for uncomplicated Plasmodium falciparum malaria in Malawi, 2014. Malar J 2016; 15:236. [PMID: 27113085 PMCID: PMC4845327 DOI: 10.1186/s12936-016-1281-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/12/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Malaria causes significant morbidity in Malawi, with an estimated 5 million cases in 2014. Artemether-lumefantrine (AL) and artesunate-amodiaquine (ASAQ) are the first- and second-line treatments for uncomplicated malaria, respectively, but emerging resistance threatens their efficacy. In order to understand whether AL and ASAQ remain efficacious for the treatment of uncomplicated Plasmodium falciparum malaria in Malawi, a therapeutic efficacy trial was conducted. METHODS During March-July 2014, febrile children aged 6-59 months with microscopy-confirmed uncomplicated P. falciparum malaria (1000-200,000 parasites/μL) were enrolled in a 28-day randomized in vivo efficacy trial at three sites: one each in northern (Karonga), central (Nkhotakota) and southern (Machinga) Malawi. The study was powered to estimate site-specific efficacy for AL and overall efficacy for ASAQ, with 3:1 randomization to AL or ASAQ. Blood was collected for malaria microscopy and molecular testing on days 0-3, 7, 14, 21, and 28. Recrudescence and reinfection were differentiated using polymerase chain reaction (PCR) genotyping of merozoite surface protein. The primary outcome was the PCR-corrected day 28 Kaplan-Meier cumulative success rate. RESULTS A total of 452 children were enrolled; 303/338 (89 %) and 98/114 (86 %) reached a study endpoint in AL and ASAQ arms, respectively. All treatment failures occurred after day 3. The day 28 uncorrected cumulative success rate was 97.1 % (95 % confidence interval [CI]: 93.9-100 %) for ASAQ and 76.8 % (95 % CI 72.1-81.5 %) for AL, with 82.5 % (95 % CI 75.4-89.7 %), 69 % (95 % CI 59.9-78.1 %), and 78.2 % (95 % CI 70.2-86.3 %) success in the northern, central, and southern regions, respectively. The day 28 PCR-corrected cumulative success rate was 99 % (95 % CI 97.2-100 %) in the ASAQ arm and 99.3 % (95 % CI 98.3-100 %) in the AL arm, with 98-100 % efficacy in each site. CONCLUSIONS As evidenced by the day 28 PCR-corrected cumulative success rates, both AL and ASAQ remain efficacious treatments for uncomplicated malaria in Malawi. The lower uncorrected efficacy in the AL arm compared to ASAQ may be explained by the shorter half-life of lumefantrine (3-6 days) compared to amodiaquine (9-18 days). The high reinfection rate suggests that there is a continued need to scale-up effective malaria prevention interventions.
Collapse
Affiliation(s)
- Magdalena Paczkowski
- />Malaria Branch, Division of Parasitic Diseases & Malaria, Center for Global Health, Centers for Disease Control and Prevention, 1600 Clifton Rd. NE, Mailstop A06, Atlanta, GA 30329-4027 USA
| | - Dyson Mwandama
- />Malaria Alert Centre, University of Malawi College of Medicine, Blantyre, Malawi
| | | | - Madalitso Luka
- />Malaria Alert Centre, University of Malawi College of Medicine, Blantyre, Malawi
| | - Georgina Makuta
- />Malaria Alert Centre, University of Malawi College of Medicine, Blantyre, Malawi
| | - John Sande
- />National Malaria Control Programme, Ministry of Health, Lilongwe, Malawi
| | - Doreen Ali
- />National Malaria Control Programme, Ministry of Health, Lilongwe, Malawi
| | - Peter Troell
- />US President’s Malaria Initiative, Malaria Branch, Division of Parasitic Diseases and Malaria, US Centers for Disease Control and Prevention, Lilongwe, Malawi
| | - Don P. Mathanga
- />Malaria Alert Centre, University of Malawi College of Medicine, Blantyre, Malawi
| | - Julie Gutman
- />Malaria Branch, Division of Parasitic Diseases & Malaria, Center for Global Health, Centers for Disease Control and Prevention, 1600 Clifton Rd. NE, Mailstop A06, Atlanta, GA 30329-4027 USA
| |
Collapse
|
50
|
Kiaco K, Teixeira J, Machado M, do Rosário V, Lopes D. Evaluation of artemether-lumefantrine efficacy in the treatment of uncomplicated malaria and its association with pfmdr1, pfatpase6 and K13-propeller polymorphisms in Luanda, Angola. Malar J 2015; 14:504. [PMID: 26670642 PMCID: PMC4681156 DOI: 10.1186/s12936-015-1018-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 11/26/2015] [Indexed: 11/10/2022] Open
Abstract
Background Drug resistance in Plasmodiumfalciparum has posed an obstacle to effective treatment and challenges many malaria control programmes in endemic areas. In Angola, until 2003, chloroquine (CQ) was used as first-line therapy for uncomplicated malaria. It was replaced initially by amodiaquine and, in 2006, by artemisinin-based combination therapy (ACT) with artemether-lumefantrine (AL, Coartem®). Efficacy study of ACT, conducted in Angola between 2004 and 2005, showed a baseline efficacy of ≈99 %. Methods 103 malaria patients were enrolled according to WHO proceedings. Patients were followed up with clinical and parasitological evaluations for 28 days, parasite density and identification was evaluated by microscopy, the pfmsp2 were genotyped by nested-PCR, to distinguish parasite recrudescence from new infections; the polymorphisms at codons 86 and 1246 of pfmdr1 gene, and 769 of pfatp6 gene were assessed by PCR–RFLP and sequencing for pfk13-propeller genotype. Results The cure rate was 91.3 %. The obtained results showed that from 103 patients, 12.6 % (n = 13) still had parasitaemia 1 day after the treatment was finished. On day 0, of the 94 evaluated samples, wild-type alleles were identified in 73.4 % (n = 69) for pfmdr1 N86Y position and only one sample carried the mutant allele (Y) for pfmdr1 1246; 14 % of these samples showed increased pfmdr1 copy number; 100 % (n = 21) had wild-type allele of k13 gene in all the studied positions. Discussion These results showed changes in parasite profile susceptibility to AL in comparison to the baseline data from 2002 to 2004 and on the genotyping characteristics; the clinical outcome after treatment with AL did not link a particular genotype with treatment failure; observed changes do not provide sufficient evidence for a treatment policy change, but they suggest that a carefully monitoring is needed in this area.
Collapse
Affiliation(s)
- Kinanga Kiaco
- Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008, Lisbon, Portugal. .,Serviços de Saúde das Forças Armadas Angolanas, Estado Maior General das Forças Armadas, Luanda, Angola.
| | - Joana Teixeira
- Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008, Lisbon, Portugal.
| | - Marta Machado
- Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008, Lisbon, Portugal. .,Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008, Lisbon, Portugal.
| | - Virgílio do Rosário
- Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008, Lisbon, Portugal.
| | - Dinora Lopes
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008, Lisbon, Portugal.
| |
Collapse
|