Parasitic Infection and Immunity

A special issue of Biomedicines (ISSN 2227-9059). This special issue belongs to the section "Molecular and Translational Medicine".

Deadline for manuscript submissions: closed (31 July 2021) | Viewed by 28126

Special Issue Editors


E-Mail Website
Guest Editor
Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal
Interests: protozoa; imunology; leishmaniasis; Chagas disease
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Escola Superior Agrária de Viseu (ESAV), Instituto Politécnico de Viseu (IPV), Viseu Portugal and Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Lisboa, Portugal
Interests: canine leishmaniasis; parasite immunology; cellular immunology; molecular immunology; innate immune response; parasite–host interaction; neutrophils; macrophages
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Emerging and reemerging parasitic infections affecting humans and animals represent a significant public health concern. The biological complexity of protozoa, helminths and ectoparasites and the specificity of host immune responses required for parasite control have compromised the development of effective therapeutic and prophylactic tools. Deciphering the complex molecular events that regulate host–parasite interaction through the application of new and sophisticated approaches—such as genomics, reverse genetic analysis, proteomics and bioinformatics techniques—could fill in some knowledge gaps. Since many parasitic diseases are zoonotic, research involving the One Health concept is critical to increase our ability to treat, control and prevent many of these diseases.

Thus, we cordially invite authors to submit original research or review articles pertaining to parasites and host–parasite interactions.

Dr. Gabriela Santos-Gomes
Dr. Maria Pereira
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Biomedicines is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • mechanisms of parasite pathogenesis
  • extracellular vesicles
  • escape mechanisms employed by parasites
  • host resistance-susceptibility
  • innate and adaptive cellular immune response
  • experimental and translational models of infection

Published Papers (11 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Other

5 pages, 208 KiB  
Editorial
Parasitic Infection and Immunity—A Special Biomedicines Issue
by Maria A. Pereira and Gabriela Santos-Gomes
Biomedicines 2022, 10(10), 2547; https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10102547 - 12 Oct 2022
Cited by 2 | Viewed by 1037
Abstract
Infectious parasitic diseases that affect humans and animals remain a central health concern worldwide [...] Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)

Research

Jump to: Editorial, Other

17 pages, 3036 KiB  
Article
African Trypanosomiasis: Extracellular Vesicles Shed by Trypanosoma brucei brucei Manipulate Host Mononuclear Cells
by Tatiana Dias-Guerreiro, Joana Palma-Marques, Patrícia Mourata-Gonçalves, Graça Alexandre-Pires, Ana Valério-Bolas, Áurea Gabriel, Telmo Nunes, Wilson Antunes, Isabel Pereira da Fonseca, Marcelo Sousa-Silva and Gabriela Santos-Gomes
Biomedicines 2021, 9(8), 1056; https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines9081056 - 20 Aug 2021
Cited by 9 | Viewed by 3503
Abstract
African trypanosomiasis or sleeping sickness is a zoonotic disease caused by Trypanosoma brucei, a protozoan parasite transmitted by Glossina spp. (tsetse fly). Parasite introduction into mammal hosts triggers a succession of events, involving both innate and adaptive immunity. Macrophages (MΦ) have a [...] Read more.
African trypanosomiasis or sleeping sickness is a zoonotic disease caused by Trypanosoma brucei, a protozoan parasite transmitted by Glossina spp. (tsetse fly). Parasite introduction into mammal hosts triggers a succession of events, involving both innate and adaptive immunity. Macrophages (MΦ) have a key role in innate defence since they are antigen-presenting cells and have a microbicidal function essential for trypanosome clearance. Adaptive immune defence is carried out by lymphocytes, especially by T cells that promote an integrated immune response. Like mammal cells, T. b. brucei parasites release extracellular vesicles (TbEVs), which carry macromolecules that can be transferred to host cells, transmitting biological information able to manipulate cell immune response. However, the exact role of TbEVs in host immune response remains poorly understood. Thus, the current study examined the effect elicited by TbEVs on MΦ and T lymphocytes. A combined approach of microscopy, nanoparticle tracking analysis, multiparametric flow cytometry, colourimetric assays and detailed statistical analyses were used to evaluate the influence of TbEVs in mouse mononuclear cells. It was shown that TbEVs can establish direct communication with cells of innate and adaptative immunity. TbEVs induce the differentiation of both M1- and M2-MΦ and elicit the expansion of MHCI+, MHCII+ and MHCI+MHCII+ MΦ subpopulations. In T lymphocytes, TbEVs drive the overexpression of cell-surface CD3 and the nuclear factor FoxP3, which lead to the differentiation of regulatory CD4+ and CD8+ T cells. Moreover, this study indicates that T. b. brucei and TbEVs seem to display opposite but complementary effects in the host, establishing a balance between parasite growth and controlled immune response, at least during the early phase of infection. Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)
Show Figures

Figure 1

18 pages, 10159 KiB  
Article
Anti-Toxoplasma gondii Effects of a Novel Spider Peptide XYP1 In Vitro and In Vivo
by Yuan Liu, Yaqin Tang, Xing Tang, Mengqi Wu, Shengjie Hou, Xiaohua Liu, Jing Li, Meichun Deng, Shuaiqin Huang and Liping Jiang
Biomedicines 2021, 9(8), 934; https://doi.org/10.3390/biomedicines9080934 - 01 Aug 2021
Cited by 11 | Viewed by 2342 | Correction
Abstract
Toxoplasmosis, caused by an obligate intracellular parasite Toxoplasma gondii, is one of the most prevalent zoonoses worldwide. Treatments for this disease by traditional drugs have shown numerous side effects, thus effective alternative anti-Toxoplasma strategies or drugs are urgently needed. In this [...] Read more.
Toxoplasmosis, caused by an obligate intracellular parasite Toxoplasma gondii, is one of the most prevalent zoonoses worldwide. Treatments for this disease by traditional drugs have shown numerous side effects, thus effective alternative anti-Toxoplasma strategies or drugs are urgently needed. In this study, a novel spider peptide, XYP1, was identified from the cDNA library of the venom gland of the spider Lycosa coelestis. Our results showed that XYP1 has potent anti-Toxoplasma activity in vitro and in vivo. Specifically, treatment with XYP1 significantly inhibited the viability, invasion and proliferation of tachyzoites with low cytotoxicity (IC50 = 38.79 μΜ) on human host cells, and increased the survival rate of mice acutely infected with T. gondii. Next, scanning electron microscopy, transmission electron microscopy and RNA sequencing were employed to further explore the functional mechanism of XYP1, and the results indicated that XYP1 causes membrane perforation, swelling and disruption of tachyzoites, which could be closely associated with differential expression of several membrane-associated proteins including HSP29. In conclusion, XYP1 may be a promising new drug candidate for the treatment of toxoplasmosis. Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)
Show Figures

Figure 1

16 pages, 3480 KiB  
Article
Unexpected Role of Sterol Synthesis in RNA Stability and Translation in Leishmania
by Zemfira N. Karamysheva, Samrat Moitra, Andrea Perez, Sumit Mukherjee, Elena B. Tikhonova, Andrey L. Karamyshev and Kai Zhang
Biomedicines 2021, 9(6), 696; https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines9060696 - 19 Jun 2021
Cited by 3 | Viewed by 2285
Abstract
Leishmania parasites are trypanosomatid protozoans that cause leishmaniasis affecting millions of people worldwide. Sterols are important components of the plasma and organellar membranes. They also serve as precursors for the synthesis of signaling molecules. Unlike animals, Leishmania does not synthesize cholesterol but makes [...] Read more.
Leishmania parasites are trypanosomatid protozoans that cause leishmaniasis affecting millions of people worldwide. Sterols are important components of the plasma and organellar membranes. They also serve as precursors for the synthesis of signaling molecules. Unlike animals, Leishmania does not synthesize cholesterol but makes ergostane-based sterols instead. C-14-demethylase is a key enzyme involved in the biosynthesis of sterols and an important drug target. In Leishmania parasites, the inactivation of C-14-demethylase leads to multiple defects, including increased plasma membrane fluidity, mitochondrion dysfunction, hypersensitivity to stress and reduced virulence. In this study, we revealed a novel role for sterol synthesis in the maintenance of RNA stability and translation. Sterol alteration in C-14-demethylase knockout mutant leads to increased RNA degradation, reduced translation and impaired heat shock response. Thus, sterol biosynthesis in Leishmania plays an unexpected role in global gene regulation. Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)
Show Figures

Figure 1

29 pages, 13637 KiB  
Article
Re-Discovery of Giardiavirus: Genomic and Functional Analysis of Viruses from Giardia duodenalis Isolates
by Gianluca Marucci, Ilaria Zullino, Lucia Bertuccini, Serena Camerini, Serena Cecchetti, Agostina Pietrantoni, Marialuisa Casella, Paolo Vatta, Alex D. Greenwood, Annarita Fiorillo and Marco Lalle
Biomedicines 2021, 9(6), 654; https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines9060654 - 08 Jun 2021
Cited by 6 | Viewed by 2578
Abstract
Giardiasis, caused by the protozoan parasite Giardia duodenalis, is an intestinal diarrheal disease affecting almost one billion people worldwide. A small endosymbiotic dsRNA viruses, G. lamblia virus (GLV), genus Giardiavirus, family Totiviridae, might inhabit human and animal isolates of G. [...] Read more.
Giardiasis, caused by the protozoan parasite Giardia duodenalis, is an intestinal diarrheal disease affecting almost one billion people worldwide. A small endosymbiotic dsRNA viruses, G. lamblia virus (GLV), genus Giardiavirus, family Totiviridae, might inhabit human and animal isolates of G. duodenalis. Three GLV genomes have been sequenced so far, and only one was intensively studied; moreover, a positive correlation between GLV and parasite virulence is yet to be proved. To understand the biological significance of GLV infection in Giardia, the characterization of several GLV strains from naturally infected G. duodenalis isolates is necessary. Here we report high-throughput sequencing of four GLVs strains, from Giardia isolates of human and animal origin. We also report on a new, unclassified viral sequence (designed GdRV-2), unrelated to Giardiavirus, encoding and expressing for a single large protein with an RdRp domain homologous to Totiviridae and Botybirnaviridae. The result of our sequencing and proteomic analyses challenge the current knowledge on GLV and strongly suggest that viral capsid protein translation unusually starts with a proline and that translation of the RNA-dependent RNA polymerase (RdRp) occurs via a +1/−2 ribosomal frameshift mechanism. Nucleotide polymorphism, confirmed by mass-spectrometry analysis, was also observed among and between GLV strains. Phylogenetic analysis indicated the occurrence of at least two GLV subtypes which display different phenotypes and transmissibility in experimental infections of a GLV naïve Giardia isolate. Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)
Show Figures

Figure 1

18 pages, 1448 KiB  
Article
Probing the Rhipicephalus bursa Sialomes in Potential Anti-Tick Vaccine Candidates: A Reverse Vaccinology Approach
by Joana Couto, Gonçalo Seixas, Christian Stutzer, Nicholas A. Olivier, Christine Maritz-Olivier, Sandra Antunes and Ana Domingos
Biomedicines 2021, 9(4), 363; https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines9040363 - 31 Mar 2021
Cited by 10 | Viewed by 2889
Abstract
In the wake of the ‘omics’ explosion of data, reverse vaccinology approaches are being applied more readily as an alternative for the discovery of candidates for next generation diagnostics and vaccines. Promising protective antigens for the control of ticks and tick-borne diseases can [...] Read more.
In the wake of the ‘omics’ explosion of data, reverse vaccinology approaches are being applied more readily as an alternative for the discovery of candidates for next generation diagnostics and vaccines. Promising protective antigens for the control of ticks and tick-borne diseases can be discovered by mining available omics data for immunogenic epitopes. The present study aims to explore the previously obtained Rhipicephalus bursa sialotranscriptome during both feeding and Babesia infection, to select antigenic targets that are either membrane-associated or a secreted protein, as well as unique to the ectoparasite and not present in the mammalian host. Further, they should be capable of stimulating T and B cells for a potential robust immune response, and be non-allergenic or toxic to the host. From the R. bursa transcriptome, 5706 and 3025 proteins were identified as belonging to the surfaceome and secretome, respectively. Following a reverse genetics immunoinformatics pipeline, nine preferred candidates, consisting of one transmembrane-related and eight secreted proteins, were identified. These candidates showed a higher predicted antigenicity than the Bm86 antigen, with no homology to mammalian hosts and exposed regions. Only four were functionally annotated and selected for further in silico analysis, which examined their protein structure, surface accessibility, flexibility, hydrophobicity, and putative linear B and T-cell epitopes. Regions with overlapping coincident epitopes groups (CEGs) were evaluated to select peptides that were further analyzed for their physicochemical characteristics, potential allergenicity, toxicity, solubility, and potential propensity for crystallization. Following these procedures, a set of three peptides from the three R. bursa proteins were selected. In silico results indicate that the designed epitopes could stimulate a protective and long-lasting immune response against those tick proteins, reflecting its potential as anti-tick vaccines. The immunogenicity of these peptides was evaluated in a pilot immunization study followed by tick feeding to evaluate its impact on tick behavior and pathogen transmission. Combining in silico methods with in vivo immunogenicity evaluation enabled the screening of vaccine candidates prior to expensive infestation studies on the definitive ovine host animals. Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)
Show Figures

Figure 1

21 pages, 4209 KiB  
Article
3D-Hepatocyte Culture Applied to Parasitology: Immune Activation of Canine Hepatic Spheroids Exposed to Leishmania infantum
by Armanda V. Rodrigues, Graça Alexandre-Pires, Ana Valério-Bolas, David Santos-Mateus, Mariana Rafael-Fernandes, Maria A. Pereira, Dário Ligeiro, Telmo Nunes, Raquel Alves-Azevedo, Marcos Santos, Isabel Pereira da Fonseca and Gabriela Santos-Gomes
Biomedicines 2020, 8(12), 628; https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines8120628 - 18 Dec 2020
Cited by 3 | Viewed by 2513
Abstract
The application of innovative three-dimensional (3D) spheroids cell culture strategy to Parasitology offers the opportunity to closely explore host–parasite interactions. Here we present a first report on the application of 3D hepatic spheroids to unravel the immune response of canine hepatocytes exposed to [...] Read more.
The application of innovative three-dimensional (3D) spheroids cell culture strategy to Parasitology offers the opportunity to closely explore host–parasite interactions. Here we present a first report on the application of 3D hepatic spheroids to unravel the immune response of canine hepatocytes exposed to Leishmania infantum. The liver, usually considered a major metabolic organ, also performs several important immunological functions and constitutes a target organ for L. infantum infection, the etiological agent of canine leishmaniasis (CanL), and a parasitic disease of major veterinary and public health concern. 3D hepatic spheroids were able to sense and immunologically react to L. infantum parasites, generating an innate immune response by increasing nitric oxide (NO) production and enhancing toll-like receptor (TLR) 2 and interleukin-10 gene expression. The immune response orchestrated by canine hepatocytes also lead to the impairment of several cytochrome P450 (CYP450) with possible implications for liver natural xenobiotic metabolization capacity. The application of meglumine antimoniate (MgA) increased the inflammatory response of 3D hepatic spheroids by inducing the expression of Nucleotide oligomerization domain (NOD) -like receptors 1 and NOD2 and TLR2, TLR4, and TLR9 and enhancing gene expression of tumour necrosis factor α. It is therefore suggested that hepatocytes are key effector cells and can activate and orchestrate the immune response to L. infantum parasites. Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)
Show Figures

Graphical abstract

17 pages, 2302 KiB  
Article
LmjF.22.0810 from Leishmania major Modulates the Th2-Type Immune Response and Is Involved in Leishmaniasis Outcome
by Andrés Vacas, Celia Fernández-Rubio, Esther Larrea, José Peña-Guerrero and Paul A. Nguewa
Biomedicines 2020, 8(11), 452; https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines8110452 - 26 Oct 2020
Cited by 9 | Viewed by 2198
Abstract
A novel serine/threonine protein kinase, LmjF.22.0810, was recently described in Leishmania major. After generating an L. major cell line overexpressing LmjF.22.0810 (named LmJ3OE), the ability of this novel protein to modulate the Th2-type immune response was analyzed. Our results suggest that the [...] Read more.
A novel serine/threonine protein kinase, LmjF.22.0810, was recently described in Leishmania major. After generating an L. major cell line overexpressing LmjF.22.0810 (named LmJ3OE), the ability of this novel protein to modulate the Th2-type immune response was analyzed. Our results suggest that the protein kinase LmjF.22.0810 might be involved in leishmaniasis outcomes. Indeed, our study outlined the LmJ3OE parasites infectivity in vitro and in vivo. Transgenic parasites displayed lower phagocytosis rates in vitro, and their promastigote forms exhibited lower expression levels of virulence factors compared to their counterparts in control parasites. In addition, LmJ3OE parasites developed significantly smaller footpad swelling in susceptible BALB/c mice. Hematoxylin–eosin staining allowed the observation of a lower inflammatory infiltrate in the footpad from LmJ3OE-infected mice compared to animals inoculated with control parasites. Gene expression of Th2-associated cytokines and effectors revealed a dramatically lower induction in interleukin (IL)-4, IL-10, and arginase 1 (ARG1) mRNA levels at the beginning of the swelling; no expression change was found in Th1-associated cytokines except for IL-12. Accordingly, such results were validated by immunohistochemistry studies, illustrating a weaker expression of ARG1 and a similar induction for inducible NO synthase (iNOS) in footpads from LmJ3OE-infected mice compared to control L. major infected animals. Furthermore, the parasite burden was lower in footpads from LmJ3OE-infected mice. Our analysis indicated that such significant smaller footpad swellings might be due to an impairment of the Th2 immune response that subsequently benefits Th1 prevalence. Altogether, these studies depict LmjF.22.0810 as a potential modulator of host immune responses to Leishmania. Finally, this promising target might be involved in the modulation of infection outcome. Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)
Show Figures

Figure 1

17 pages, 4447 KiB  
Article
S-Methylcysteine (SMC) Ameliorates Intestinal, Hepatic, and Splenic Damage Induced by Cryptosporidium parvum Infection Via Targeting Inflammatory Modulators and Oxidative Stress in Swiss Albino Mice
by Ehab Kotb Elmahallawy, Gehad E. Elshopakey, Amira A. Saleh, Ahmad Agil, Ahmed El-Morsey, Dina M. M. EL-shewehy, Ahmed S. Sad, Tokuma Yanai and Walied Abdo
Biomedicines 2020, 8(10), 423; https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines8100423 - 15 Oct 2020
Cited by 18 | Viewed by 3339
Abstract
Cryptosporidiosis has been proposed to be one of the major causes of diarrhoeal disease in humans worldwide that possesses zoonotic concern. Thereby, this study investigated the potential effects of s-Methylcysteine (SMC) on the parasite in vivo followed by the measurement of cytokines, oxidative [...] Read more.
Cryptosporidiosis has been proposed to be one of the major causes of diarrhoeal disease in humans worldwide that possesses zoonotic concern. Thereby, this study investigated the potential effects of s-Methylcysteine (SMC) on the parasite in vivo followed by the measurement of cytokines, oxidative stress parameters, and an investigation of the major histopathological changes. Sixty male Swiss albino mice weighing 20–25 g were allocated equally into five groups and orally administered saline only (control), SMC only (SMC50) (50 mg/kg b.w.), and 104Cryptosporidium parvum oocysts per mouse via an esophageal tube (C + ve untreated). The fourth and fifth groups (C + SMC25, C + SMC50) administrated 104C. parvum oocysts combined with SMC25 (low dose) and 50 (high dose) mg/kg b.w., respectively. At days 7 and 14 post-infection (PI), the feces was collected from each group in order to count C. parvum oocysts. After two weeks of treatment, the animals were euthanized and the serum was collected for biochemical analysis. Next, the intestinal, spleen, and liver sections were dissected for histopathological examination. The results revealed lower oocyst numbers in the C + SMC25 and C + SMC50 groups compared to the infected untreated group. Moreover, higher doses of SMC treatment significantly reduced the enteritis induced by C. parvum in a dose-dependent manner. The hepatic lesions were also mitigated as demonstrated in C + SMC25 and C + SMC50 groups unlike the infected group via lowering the serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP) enzymes and increasing albumin and globulin serum levels. SMC administration also reduced cytokines production (SAP, TNF-α, IL-6, and IFN-γ) mediated by Cryptosporidium infection in contrast to the infected untreated group. There were marked lymphoid depletion and amyloidosis observed in the infected untreated group, while the treated groups showed obvious increase in the lymphoid elements. Moreover, the scoring of intestinal parasites, hepatic, and splenic lesions in the SMC-treated groups exhibited significantly lower pathological lesions in different organs in a dose-dependent manner, compared to the infected untreated group. Our results also revealed a significant change in the malondialdehyde content with an elevation of glutathione and superoxide dismutase in the intestines collected from C + SMC25 and C + SMC50 mice relative to the untreated group. Taken together, our results indicated that SMC could be a promising effective compound for treating and declining C. parvum infestation via restoring structural alterations in different tissues, enhancing antioxidant enzymes, and suppressing the cytokines liberation. Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)
Show Figures

Figure 1

13 pages, 1707 KiB  
Article
Organoarsenic Compounds with In Vitro Activity against the Malaria Parasite Plasmodium falciparum
by Sofia Basova, Nathalie Wilke, Jan Christoph Koch, Aram Prokop, Albrecht Berkessel, Gabriele Pradel and Che Julius Ngwa
Biomedicines 2020, 8(8), 260; https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines8080260 - 02 Aug 2020
Cited by 8 | Viewed by 2898
Abstract
The rapid development of parasite drug resistance as well as the lack of medications targeting both the asexual and the sexual blood stages of the malaria parasite necessitate the search for novel antimalarial compounds. Eleven organoarsenic compounds were synthesized and tested for their [...] Read more.
The rapid development of parasite drug resistance as well as the lack of medications targeting both the asexual and the sexual blood stages of the malaria parasite necessitate the search for novel antimalarial compounds. Eleven organoarsenic compounds were synthesized and tested for their effect on the asexual blood stages and sexual transmission stages of the malaria parasite Plasmodium falciparum using in vitro assays. The inhibitory potential of the compounds on blood stage viability was tested on the chloroquine (CQ)-sensitive 3D7 and the CQ-resistant Dd2 strain using the Malstat assay. The most effective compounds were subsequently investigated for their effect on impairing gametocyte development and gametogenesis, using the gametocyte-producing NF54 strain in respective cell-based assays. Their potential toxicity was investigated on leukemia cell line Nalm-6 and non-infected erythrocytes. Five out of the 11 compounds showed antiplasmodial activities against 3D7, with half-maximal inhibitory concentration (IC50) values ranging between 1.52 and 8.64 µM. Three of the compounds also acted against Dd2, with the most active compound As-8 exhibiting an IC50 of 0.35 µM. The five compounds also showed significant inhibitory effects on the parasite sexual stages at both IC50 and IC90 concentrations with As-8 displaying the best gametocytocidal activity. No hemolytic and cytotoxic effect was observed for any of the compounds. The organoarsenic compound As-8 may represent a good lead for the design of novel organoarsenic drugs with combined antimalarial and transmission blocking activities. Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)
Show Figures

Figure 1

Other

Jump to: Editorial, Research

3 pages, 7312 KiB  
Correction
Correction: Liu et al. Anti-Toxoplasma gondii Effects of a Novel Spider Peptide XYP1 In Vitro and In Vivo. Biomedicines 2021, 9, 934
by Yuan Liu, Yaqin Tang, Xing Tang, Mengqi Wu, Shengjie Hou, Xiaohua Liu, Jing Li, Meichun Deng, Shuaiqin Huang and Liping Jiang
Biomedicines 2022, 10(5), 1176; https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10051176 - 19 May 2022
Viewed by 1039
Abstract
In the original publication, there were mistakes in Figure 3C, Figure 6B and Figure S2A,B as published [...] Full article
(This article belongs to the Special Issue Parasitic Infection and Immunity)
Show Figures

Figure 3

Back to TopTop