1
|
Balakrishnan A, Hunziker M, Tiwary P, Pandey V, Drew D, Billker O. A CRISPR homing screen finds a chloroquine resistance transporter-like protein of the Plasmodium oocyst essential for mosquito transmission of malaria. Nat Commun 2025; 16:3895. [PMID: 40274854 PMCID: PMC12022033 DOI: 10.1038/s41467-025-59099-1] [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: 06/11/2024] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Genetic screens with barcoded PlasmoGEM vectors have identified thousands of Plasmodium berghei gene functions in haploid blood stages, gametocytes and liver stages. However, the formation of diploid cells by fertilisation has hindered similar research on the parasites' mosquito stages. In this study, we develop a scalable genetic system that uses barcoded gene targeting vectors equipped with a CRISPR-mediated homing mechanism to generate homozygous loss-of-function mutants after one parent introduces a modified allele into the zygote. To achieve this, we use vectors additionally expressing a target gene specific gRNA. When integrated into one of the parental alleles it directs Cas9 to the intact allele after fertilisation, leading to its disruption. This homing strategy is 90% effective at generating homozygous gene editing of a fluorescence-tagged reporter locus in the oocyst. A pilot screen identifies PBANKA_0916000 as a chloroquine resistance transporter-like protein (CRTL) essential for oocyst growth and sporogony, pointing to an unexpected importance for malaria transmission of the poorly understood digestive vacuole of the oocyst that contains hemozoin granules. Homing screens provide a method for the systematic discovery of malaria transmission genes whose first essential functions are after fertilisation in the bloodmeal, enabling their potential as targets for transmission-blocking interventions to be assessed.
Collapse
Affiliation(s)
- Arjun Balakrishnan
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Mirjam Hunziker
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Puja Tiwary
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Vikash Pandey
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - David Drew
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Oliver Billker
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden.
- Department of Molecular Biology, Umeå University, Umeå, Sweden.
- Umea Centre for Microbial Research, Umeå University, Umeå, Sweden.
| |
Collapse
|
2
|
García-Guerrero AE, Marvin RG, Blackwell AM, Sigala PA. Biogenesis of Cytochromes c and c1 in the Electron Transport Chain of Malaria Parasites. ACS Infect Dis 2025; 11:813-826. [PMID: 39481007 PMCID: PMC11991887 DOI: 10.1021/acsinfecdis.4c00450] [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] [Indexed: 11/02/2024]
Abstract
Plasmodium malaria parasites retain an essential mitochondrional electron transport chain (ETC) that is critical for growth within humans and mosquitoes and is a key antimalarial drug target. ETC function requires cytochromes c and c1, which are unusual among heme proteins due to their covalent binding to heme via conserved CXXCH sequence motifs. Heme attachment to these proteins in most eukaryotes requires the mitochondrial enzyme holocytochrome c synthase (HCCS) that binds heme and the apo cytochrome to facilitate the biogenesis of the mature cytochrome c or c1. Although humans encode a single bifunctional HCCS that attaches heme to both proteins, Plasmodium parasites are like yeast and encode two separate HCCS homologues thought to be specific for heme attachment to cyt c (HCCS) or cyt c1 (HCC1S). To test the function and specificity of Plasmodium falciparum HCCS and HCC1S, we used CRISPR/Cas9 to tag both genes for conditional expression. HCC1S knockdown selectively impaired cyt c1 biogenesis and caused lethal ETC dysfunction that was not reversed by the overexpression of HCCS. Knockdown of HCCS caused a more modest growth defect but strongly sensitized parasites to mitochondrial depolarization by proguanil, revealing key defects in ETC function. These results and prior heterologous studies in Escherichia coli of cyt c hemylation by P. falciparum HCCS and HCC1S strongly suggest that both homologues are essential for mitochondrial ETC function and have distinct specificities for the biogenesis of cyt c and c1, respectively, in parasites. This study lays a foundation to develop novel strategies to selectively block ETC function in malaria parasites.
Collapse
Affiliation(s)
- Aldo E. García-Guerrero
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA 84112
| | - Rebecca G. Marvin
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA 84112
| | - Amanda Mixon Blackwell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA 84112
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA 84112
| |
Collapse
|
3
|
Jang WS, Choi MK, Choe YL, Lim CS. Development of a rapid malaria LAMP-MS assay for diagnosis of malaria infections. Sci Rep 2025; 15:8547. [PMID: 40074752 PMCID: PMC11904186 DOI: 10.1038/s41598-025-92935-4] [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: 12/30/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Malaria remains a critical global health concern, especially in tropical and subtropical regions, where it causes substantial morbidity and mortality. Current diagnostic methods, such as microscopy and PCR-based assays, are reliable but often impractical in resource-limited settings due to their dependency on complex equipment and skilled personnel. This study developed a novel malaria diagnostic platform by combining the Chelex-100/boiling DNA extraction method with a Loop-mediated Isothermal Amplification-MicroScanner (LAMP-MS) assay. The Chelex-100/boiling method is simpler and more cost-effective than conventional DNA extraction processes, making it suitable for use in resource-limited settings. The LAMP-MS assay enables multiplex detection through a microchip design with four chambers. Each chamber of the microchip is preloaded with specific primers targeting Pan, Plasmodium falciparum (Pf), Plasmodium vivax (Pv), and an internal control, minimizing non-specific amplification in multiplex LAMP reactions. In a clinical evaluation of 260 samples, the assay demonstrated a sensitivity of 97.5% for the Pan target and 100% for the Pf-specific target in the 80 Plasmodium falciparum (Pf) clinical samples. Similarly, for the 80 Plasmodium vivax (Pv) clinical samples, the assay achieved a sensitivity of 95% for the Pan target and 94% for the Pv-specific target. Notably, in the 100 non-infected clinical samples, the assay exhibited 100% specificity, with no false positives observed. These findings suggest that LAMP-MS is a rapid and reliable alternative to PCR-based methods, especially in resource-limited environments.
Collapse
Affiliation(s)
- Woong Sik Jang
- Emergency Medicine, College of Medicine, Korea University Guro Hospital, 148, Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Min Kyeong Choi
- BK21 Graduate Program, Department of Biomedical Sciences, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Young Lan Choe
- Department of Laboratory Medicine, College of Medicine, Korea University Guro Hospital, 148, Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Chae Seung Lim
- Department of Laboratory Medicine, College of Medicine, Korea University Guro Hospital, 148, Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea.
| |
Collapse
|
4
|
Rios KT, McGee JP, Sebastian A, Gedara SA, Moritz RL, Feric M, Absalon S, Swearingen KE, Lindner SE. Widespread release of translational repression across Plasmodium's host-to-vector transmission event. PLoS Pathog 2025; 21:e1012823. [PMID: 39777415 PMCID: PMC11750109 DOI: 10.1371/journal.ppat.1012823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/21/2025] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Malaria parasites must respond quickly to environmental changes, including during their transmission between mammalian and mosquito hosts. Therefore, female gametocytes proactively produce and translationally repress mRNAs that encode essential proteins that the zygote requires to establish a new infection. While the release of translational repression of individual mRNAs has been documented, the details of the global release of translational repression have not. Moreover, changes in the spatial arrangement and composition of the DOZI/CITH/ALBA complex that contribute to translational control are also not known. Therefore, we have conducted the first quantitative, comparative transcriptomics and DIA-MS proteomics of Plasmodium parasites across the host-to-vector transmission event to document the global release of translational repression. Using female gametocytes and zygotes of P. yoelii, we found that ~200 transcripts are released for translation soon after fertilization, including those encoding essential functions. Moreover, we identified that many transcripts remain repressed beyond this point. TurboID-based proximity proteomics of the DOZI/CITH/ALBA regulatory complex revealed substantial spatial and/or compositional changes across this transmission event, which are consistent with recent, paradigm-shifting models of translational control. Together, these data provide a model for the essential translational control mechanisms that promote Plasmodium's efficient transmission from mammalian host to mosquito vector.
Collapse
Affiliation(s)
- Kelly T. Rios
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - James P. McGee
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Aswathy Sebastian
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Sanjaya Aththawala Gedara
- Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Robert L. Moritz
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Marina Feric
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Sabrina Absalon
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | | | - Scott E. Lindner
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
5
|
Sayers C, Pandey V, Balakrishnan A, Michie K, Svedberg D, Hunziker M, Pardo M, Choudhary J, Berntsson R, Billker O. Systematic screens for fertility genes essential for malaria parasite transmission reveal conserved aspects of sex in a divergent eukaryote. Cell Syst 2024; 15:1075-1091.e6. [PMID: 39541984 DOI: 10.1016/j.cels.2024.10.008] [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: 02/09/2024] [Revised: 10/06/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
Sexual reproduction in malaria parasites is essential for their transmission to mosquitoes and offers a divergent eukaryote model to understand the evolution of sex. Through a panel of genetic screens in Plasmodium berghei, we identify 348 sex and transmission-related genes and define roles for unstudied genes as putative targets for transmission-blocking interventions. The functional data provide a deeper understanding of female metabolic reprogramming, meiosis, and the axoneme. We identify a complex of a SUN domain protein (SUN1) and a putative allantoicase (ALLC1) that is essential for male fertility by linking the microtubule organizing center to the nuclear envelope and enabling mitotic spindle formation during male gametogenesis. Both proteins have orthologs in mouse testis, and the data raise the possibility of an ancient role for atypical SUN domain proteins in coupling the nucleus and axoneme. Altogether, our data provide an unbiased picture of the molecular processes that underpin malaria parasite transmission. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Claire Sayers
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden; Department of Molecular Biology, Umeå University, Umeå, Sweden; School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Vikash Pandey
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden; Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Arjun Balakrishnan
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden; Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Katharine Michie
- Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, Australia
| | - Dennis Svedberg
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Mirjam Hunziker
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden; Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Mercedes Pardo
- Institute of Cancer Research, Chester Beatty Laboratories, London, UK
| | - Jyoti Choudhary
- Institute of Cancer Research, Chester Beatty Laboratories, London, UK
| | - Ronnie Berntsson
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Oliver Billker
- The Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden; Department of Molecular Biology, Umeå University, Umeå, Sweden.
| |
Collapse
|
6
|
Yan Y, Cheung E, Verzier LH, Appetecchia F, March S, Craven AR, Du E, Probst AS, Rinvee TA, de Vries LE, Kauffman J, Bhatia SN, Nelson E, Singh N, Peng D, Shaw WR, Catteruccia F. Mapping Plasmodium transitions and interactions in the Anopheles female. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623125. [PMID: 39605504 PMCID: PMC11601300 DOI: 10.1101/2024.11.12.623125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The human malaria parasite, Plasmodium falciparum , relies on Anopheles mosquitoes for transmission. Once ingested during blood feeding, most parasites die in the mosquito midgut lumen or during epithelium traversal. How surviving ookinetes interact with midgut cells and form oocysts is unknown, yet these steps are essential to initiate a remarkable, similarly uncharacterized growth process culminating in the production of thousands of infectious sporozoites. Here, using single-cell RNA sequencing of both parasites and mosquito cells across four time points and two metabolic conditions, we unveil key processes shaping developmental transitions and mosquito-parasite interactions occurring in the midgut. In depth functional analyses reveal processes regulating oocyst growth and identify the transcription factor Pf SIP2 as essential for sporozoite infection of human hepatocytes. By combining the analysis of shared mosquito-parasite barcodes with confocal microscopy, we discover that parasites preferentially interact with midgut progenitor cells during epithelial crossing, potentially using their basal location as an exit landmark. Additionally, we unveil tight connections between extracellular late oocysts and surrounding muscle cells that may ensure parasites adhere to the midgut without damaging it. Ultimately, our study provides fundamental insight into the molecular events characterizing previously inaccessible biological transitions and mosquito-parasite interactions, and identifies candidates for transmission-blocking strategies.
Collapse
|
7
|
García-Guerrero AE, Marvin RG, Blackwell AM, Sigala PA. Biogenesis of cytochromes c and c 1 in the electron transport chain of malaria parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.575742. [PMID: 38352463 PMCID: PMC10862854 DOI: 10.1101/2024.02.01.575742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Plasmodium malaria parasites retain an essential mitochondrional electron transport chain (ETC) that is critical for growth within humans and mosquitoes and a key antimalarial drug target. ETC function requires cytochromes c and c 1 that are unusual among heme proteins due to their covalent binding to heme via conserved CXXCH sequence motifs. Heme attachment to these proteins in most eukaryotes requires the mitochondrial enzyme holocytochrome c synthase (HCCS) that binds heme and the apo cytochrome to facilitate biogenesis of the mature cytochrome c or c 1. Although humans encode a single bifunctional HCCS that attaches heme to both proteins, Plasmodium parasites are like yeast and encode two separate HCCS homologs thought to be specific for heme attachment to cyt c (HCCS) or cyt c 1 (HCC1S). To test the function and specificity of P. falciparum HCCS and HCC1S, we used CRISPR/Cas9 to tag both genes for conditional expression. HCC1S knockdown selectively impaired cyt c 1 biogenesis and caused lethal ETC dysfunction that was not reversed by over-expression of HCCS. Knockdown of HCCS caused a more modest growth defect but strongly sensitized parasites to mitochondrial depolarization by proguanil, revealing key defects in ETC function. These results and prior heterologous studies in E. coli of cyt c hemylation by P. falciparum HCCS and HCC1S strongly suggest that both homologs are essential for mitochondrial ETC function and have distinct specificities for biogenesis of cyt c and c 1, respectively, in parasites. This study lays a foundation to develop novel strategies to selectively block ETC function in malaria parasites.
Collapse
Affiliation(s)
- Aldo E. García-Guerrero
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA 84112
| | - Rebecca G. Marvin
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA 84112
| | - Amanda Mixon Blackwell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA 84112
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA 84112
| |
Collapse
|
8
|
Cox A, Krishnankutty N, Shave S, Howick VM, Auer M, La Clair JJ, Philip N. Repositioning Brusatol as a Transmission Blocker of Malaria Parasites. ACS Infect Dis 2024; 10:3586-3596. [PMID: 39352879 PMCID: PMC11474950 DOI: 10.1021/acsinfecdis.4c00434] [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: 05/27/2024] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Currently, primaquine is the only malaria transmission-blocking drug recommended by the WHO. Recent efforts have highlighted the importance of discovering new agents that regulate malarial transmission, with particular interest in agents that can be administered in a single low dose, ideally with a discrete and Plasmodium-selective mechanism of action. Here, our team demonstrates an approach to identify malaria transmission-blocking agents through a combination of in vitro screening and in vivo analyses. Using a panel of natural products, our approach identified potent transmission blockers, as illustrated by the discovery of the transmission-blocking efficacy of brusatol. As a member of a large family of biologically active natural products, this discovery provides a critical next step toward developing methods to rapidly identify quassinoids and related agents with valuable pharmacological therapeutic properties.
Collapse
Affiliation(s)
- Amelia Cox
- School
of Biodiversity, One Health and Veterinary Medicine, College of Medical,
Veterinary and Life Sciences, University
of Glasgow, Garscube
Campus, Bearsden Road, Glasgow G61 1QH, United Kingdom
| | - Neelima Krishnankutty
- Institute
of Immunology and Infection Research, University
of Edinburgh, Ashworth Laboratories 2, Room 3.11, Edinburgh EH9 3FL, United Kingdom
| | - Steven Shave
- School
of Biological Sciences, University of Edinburgh, The King’s Buildings, Edinburgh EH9 3BF, United Kingdom
| | - Virginia M. Howick
- School
of Biodiversity, One Health and Veterinary Medicine, College of Medical,
Veterinary and Life Sciences, University
of Glasgow, Garscube
Campus, Bearsden Road, Glasgow G61 1QH, United Kingdom
| | - Manfred Auer
- School
of Biological Sciences, University of Edinburgh, The King’s Buildings, Edinburgh EH9 3BF, United Kingdom
- Xenobe
Research Institute, P.O. Box 3052, San Diego, California 92163, United States
| | - James J. La Clair
- Xenobe
Research Institute, P.O. Box 3052, San Diego, California 92163, United States
| | - Nisha Philip
- Institute
of Immunology and Infection Research, University
of Edinburgh, Ashworth Laboratories 2, Room 3.11, Edinburgh EH9 3FL, United Kingdom
| |
Collapse
|
9
|
Niu G, Wang X, Gao W, Cui L, Li J. Leucinostatins from fungal extracts block malaria transmission to mosquitoes. Parasit Vectors 2024; 17:401. [PMID: 39304934 DOI: 10.1186/s13071-024-06450-y] [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: 03/30/2024] [Accepted: 08/13/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Malaria is a mosquito-transmitted disease that kills more than half a million people annually. The lack of effective malaria vaccines and recently increasing malaria cases urge innovative approaches to prevent malaria. Previously, we reported that the extract from the soil-dwelling fungus Purpureocillium lilacinum, a common fungus from the soil, reduced Plasmodium falciparum oocysts in Anopheles gambiae midguts after mosquitoes contacted the treated surface before feeding. METHODS We used liquid chromatography to fraction fungal crude extract and tract the active fraction using a contact-wise approach and standard membrane feeding assays. The purified small molecules were analyzed using precise mass spectrometry and tandem mass spectrometry. RESULTS We isolated four active small molecules from P. lilacinum and determined them as leucinostatin A, B, A2, and B2. Pre-exposure of mosquitoes via contact with very low-concentration leucinostatin A significantly reduced the number of oocysts. The half-maximal response or inhibition concentration (EC50) via pre-exposure was 0.7 mg/m2, similar to atovaquone but lower than other known antimalarials. The inhibitory effect of leucinostatin A against P. falciparum during intraerythrocytic development, gametogenesis, sporogonic development, and ookinete formation, with the exception of oocyst development, suggests that leucinostatins play a part during parasite invasion of new cells. CONCLUSIONS Leucinostatins, secondary metabolites from P. lilacinum disrupt malaria development, particular transmission to mosquitoes by contact. The contact-wise malaria control as a nonconventional approach is highly needed in malaria-endemic areas.
Collapse
Affiliation(s)
- Guodong Niu
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | - Xiaohong Wang
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | - Wenda Gao
- Antagen Pharmaceuticals, Canton, MA, 02021, USA
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Jun Li
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
10
|
Attai K, Ekpenyong M, Amannah C, Asuquo D, Ajuga P, Obot O, Johnson E, John A, Maduka O, Akwaowo C, Uzoka FM. Enhancing the Interpretability of Malaria and Typhoid Diagnosis with Explainable AI and Large Language Models. Trop Med Infect Dis 2024; 9:216. [PMID: 39330905 PMCID: PMC11436130 DOI: 10.3390/tropicalmed9090216] [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: 08/07/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
Malaria and Typhoid fever are prevalent diseases in tropical regions, and both are exacerbated by unclear protocols, drug resistance, and environmental factors. Prompt and accurate diagnosis is crucial to improve accessibility and reduce mortality rates. Traditional diagnosis methods cannot effectively capture the complexities of these diseases due to the presence of similar symptoms. Although machine learning (ML) models offer accurate predictions, they operate as "black boxes" with non-interpretable decision-making processes, making it challenging for healthcare providers to comprehend how the conclusions are reached. This study employs explainable AI (XAI) models such as Local Interpretable Model-agnostic Explanations (LIME), and Large Language Models (LLMs) like GPT to clarify diagnostic results for healthcare workers, building trust and transparency in medical diagnostics by describing which symptoms had the greatest impact on the model's decisions and providing clear, understandable explanations. The models were implemented on Google Colab and Visual Studio Code because of their rich libraries and extensions. Results showed that the Random Forest model outperformed the other tested models; in addition, important features were identified with the LIME plots while ChatGPT 3.5 had a comparative advantage over other LLMs. The study integrates RF, LIME, and GPT in building a mobile app to enhance the interpretability and transparency in malaria and typhoid diagnosis system. Despite its promising results, the system's performance is constrained by the quality of the dataset. Additionally, while LIME and GPT improve transparency, they may introduce complexities in real-time deployment due to computational demands and the need for internet service to maintain relevance and accuracy. The findings suggest that AI-driven diagnostic systems can significantly enhance healthcare delivery in environments with limited resources, and future works can explore the applicability of this framework to other medical conditions and datasets.
Collapse
Affiliation(s)
- Kingsley Attai
- Department of Mathematics and Computer Science, Ritman University, Ikot Ekpene 530101, Nigeria; (E.J.); (A.J.)
| | - Moses Ekpenyong
- Department of Computer Science, Faculty of Computing, University of Uyo, Uyo 520103, Nigeria; (M.E.); (O.O.)
- Science, Technology, Engineering and Mathematics (STEM) Centre, University of Uyo and Centre for Research, University of Uyo, Uyo 520103, Nigeria
| | - Constance Amannah
- Department of Computer Science, Ignatius Ajuru University of Education, Port Harcourt 500102, Nigeria;
| | - Daniel Asuquo
- Department of Information Systems, Faculty of Computing, University of Uyo, Uyo 520103, Nigeria;
| | - Peterben Ajuga
- Department of Computer Engineering, Faculty of Engineering, Gregory University, Uturu 441106, Nigeria;
| | - Okure Obot
- Department of Computer Science, Faculty of Computing, University of Uyo, Uyo 520103, Nigeria; (M.E.); (O.O.)
| | - Ekemini Johnson
- Department of Mathematics and Computer Science, Ritman University, Ikot Ekpene 530101, Nigeria; (E.J.); (A.J.)
| | - Anietie John
- Department of Mathematics and Computer Science, Ritman University, Ikot Ekpene 530101, Nigeria; (E.J.); (A.J.)
| | - Omosivie Maduka
- University of Port Harcourt Teaching Hospital, Port Harcourt 500102, Nigeria;
| | | | - Faith-Michael Uzoka
- Department of Mathematics and Computing, Mount Royal University, Calgary, AB T3E 6K6, Canada;
| |
Collapse
|
11
|
Daniel L, Karam A, Franco CHJ, Conde C, Sacramento de Morais A, Mosnier J, Fonta I, Villarreal W, Pradines B, Moreira DRM, Navarro M. Metal(triphenylphosphine)-atovaquone Complexes: Synthesis, Antimalarial Activity, and Suppression of Heme Detoxification. Inorg Chem 2024; 63:17087-17099. [PMID: 39185932 PMCID: PMC11409218 DOI: 10.1021/acs.inorgchem.4c02751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
To ascertain the bioinorganic chemistry of metals conjugated with quinones, the complexes [Ag(ATV)(PPh3)2] (1), [Au(ATV)(PPh3)]·2H2O (2), and [Cu(ATV)(PPh3)2] (3) were synthesized by the coordination of the antimalarial naphthoquinone atovaquone (ATV) to the starting materials [Ag(PPh3)2]NO3, [Au(PPh3)Cl], and [Cu(PPh3)2NO3], respectively. These complexes were characterized by analytical and spectroscopical techniques. X-ray diffraction of single crystals precisely confirmed the coordination mode of ATV to the metals, which was monodentate or bidentate, depending on the metal center. Both coordination modes showed high stability in the solid state and in solution. All three complexes showed negative log D values at pH 5, but at pH 7.4, while complex 2 continued to have a negative log D value, complexes 1 and 3 displayed positive values, indicating a more hydrophilic character. ATV and complexes 1-3 could bind to ferriprotoporphyrin IX (FePPIX); however, only complexes 1-3 could inhibit β-hematin crystal formation. Phenotype-based activity revealed that all three metal complexes are able to inhibit the growth of P. falciparum with potency and selectivity comparable to those of ATV, while the starting materials lack this activity. The outcomes of this chemical design may provide significant insights into structure-activity relationships for the development of new antimalarial agents.
Collapse
Affiliation(s)
- Luana Daniel
- Laboratório de Química Bioinorgânica e Catalise, Departamento Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36036-900, Brazil
| | - Arquímedes Karam
- Laboratório de Química Bioinorgânica e Catalise, Departamento Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36036-900, Brazil
| | - Chris Hebert J Franco
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, 1049-001, Portugal
| | - Camila Conde
- Laboratório de Química Bioinorgânica e Catalise, Departamento Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36036-900, Brazil
| | | | - Joel Mosnier
- Unité Parasitologie et Entomologie, Institut de Recherche Biomédicale des Armées, Marseille, 13005, France
- Aix-Marseille Univ, SSA, AP-HM, RITMES, Marseille, 13005, France
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, Marseille, 13005, France
- Centre National de Référence du Paludisme, Marseille, 13005, France
| | - Isabelle Fonta
- Unité Parasitologie et Entomologie, Institut de Recherche Biomédicale des Armées, Marseille, 13005, France
- Aix-Marseille Univ, SSA, AP-HM, RITMES, Marseille, 13005, France
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, Marseille, 13005, France
- Centre National de Référence du Paludisme, Marseille, 13005, France
| | - Wilmer Villarreal
- Grupo de Química Inorgânica Medicinal e Reações Aplicadas, Instituto de Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 91501-970, Brazil
| | - Bruno Pradines
- Unité Parasitologie et Entomologie, Institut de Recherche Biomédicale des Armées, Marseille, 13005, France
- Aix-Marseille Univ, SSA, AP-HM, RITMES, Marseille, 13005, France
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, Marseille, 13005, France
- Centre National de Référence du Paludisme, Marseille, 13005, France
| | | | - Maribel Navarro
- Laboratório de Química Bioinorgânica e Catalise, Departamento Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36036-900, Brazil
| |
Collapse
|
12
|
Sheokand PK, Pradhan S, Maclean AE, Mühleip A, Sheiner L. Plasmodium falciparum Mitochondrial Complex III, the Target of Atovaquone, Is Essential for Progression to the Transmissible Sexual Stages. Int J Mol Sci 2024; 25:9239. [PMID: 39273187 PMCID: PMC11394760 DOI: 10.3390/ijms25179239] [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: 05/07/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/15/2024] Open
Abstract
The Plasmodium falciparum mitochondrial electron transport chain (mETC) is responsible for essential metabolic pathways such as de novo pyrimidine synthesis and ATP synthesis. The mETC complex III (cytochrome bc1 complex) is responsible for transferring electrons from ubiquinol to cytochrome c and generating a proton gradient across the inner mitochondrial membrane, which is necessary for the function of ATP synthase. Recent studies have revealed that the composition of Plasmodium falciparum complex III (PfCIII) is divergent from humans, highlighting its suitability as a target for specific inhibition. Indeed, PfCIII is the target of the clinically used anti-malarial atovaquone and of several inhibitors undergoing pre-clinical trials, yet its role in parasite biology has not been thoroughly studied. We provide evidence that the universally conserved subunit, PfRieske, and the new parasite subunit, PfC3AP2, are part of PfCIII, with the latter providing support for the prediction of its divergent composition. Using inducible depletion, we show that PfRieske, and therefore, PfCIII as a whole, is essential for asexual blood stage parasite survival, in line with previous observations. We further found that depletion of PfRieske results in gametocyte maturation defects. These phenotypes are linked to defects in mitochondrial functions upon PfRieske depletion, including increased sensitivity to mETC inhibitors in asexual stages and decreased cristae abundance alongside abnormal mitochondrial morphology in gametocytes. This is the first study that explores the direct role of the PfCIII in gametogenesis via genetic disruption, paving the way for a better understanding of the role of mETC in the complex life cycle of these important parasites and providing further support for the focus of antimalarial drug development on this pathway.
Collapse
Affiliation(s)
- Pradeep Kumar Sheokand
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, UK
| | - Sabyasachi Pradhan
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, UK
| | - Andrew E Maclean
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, UK
| | - Alexander Mühleip
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, UK
| | - Lilach Sheiner
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
13
|
Appetecchia F, Fabbrizi E, Fiorentino F, Consalvi S, Biava M, Poce G, Rotili D. Transmission-Blocking Strategies for Malaria Eradication: Recent Advances in Small-Molecule Drug Development. Pharmaceuticals (Basel) 2024; 17:962. [PMID: 39065810 PMCID: PMC11279868 DOI: 10.3390/ph17070962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Malaria drug research and development efforts have resurged in the last decade following the decelerating rate of mortality and malaria cases in endemic regions. The inefficiency of malaria interventions is largely driven by the spreading resistance of the Plasmodium falciparum parasite to current drug regimens and that of the malaria vector, the Anopheles mosquito, to insecticides. In response to the new eradication agenda, drugs that act by breaking the malaria transmission cycle (transmission-blocking drugs), which has been recognized as an important and additional target for intervention, are being developed. These drugs take advantage of the susceptibility of Plasmodium during population bottlenecks before transmission (gametocytes) and in the mosquito vector (gametes, zygotes, ookinetes, oocysts, sporozoites). To date, compounds targeting stage V gametocytes predominate in the chemical library of transmission-blocking drugs, and some of them have entered clinical trials. The targeting of Plasmodium mosquito stages has recently renewed interest in the development of innovative malaria control tools, which hold promise for the application of compounds effective at these stages. In this review, we highlight the major achievements and provide an update on the research of transmission-blocking drugs, with a particular focus on their chemical scaffolds, antiplasmodial activity, and transmission-blocking potential.
Collapse
Affiliation(s)
| | | | | | | | | | - Giovanna Poce
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| |
Collapse
|
14
|
Rios KT, McGee JP, Sebastian A, Moritz RL, Feric M, Absalon S, Swearingen KE, Lindner SE. Global Release of Translational Repression Across Plasmodium's Host-to-Vector Transmission Event. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.577866. [PMID: 38352447 PMCID: PMC10862809 DOI: 10.1101/2024.02.01.577866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Malaria parasites must be able to respond quickly to changes in their environment, including during their transmission between mammalian hosts and mosquito vectors. Therefore, before transmission, female gametocytes proactively produce and translationally repress mRNAs that encode essential proteins that the zygote requires to establish a new infection. This essential regulatory control requires the orthologues of DDX6 (DOZI), LSM14a (CITH), and ALBA proteins to form a translationally repressive complex in female gametocytes that associates with many of the affected mRNAs. However, while the release of translational repression of individual mRNAs has been documented, the details of the global release of translational repression have not. Moreover, the changes in spatial arrangement and composition of the DOZI/CITH/ALBA complex that contribute to translational control are also not known. Therefore, we have conducted the first quantitative, comparative transcriptomics and DIA-MS proteomics of Plasmodium parasites across the host-to-vector transmission event to document the global release of translational repression. Using female gametocytes and zygotes of P. yoelii, we found that nearly 200 transcripts are released for translation soon after fertilization, including those with essential functions for the zygote. However, we also observed that some transcripts remain repressed beyond this point. In addition, we have used TurboID-based proximity proteomics to interrogate the spatial and compositional changes in the DOZI/CITH/ALBA complex across this transmission event. Consistent with recent models of translational control, proteins that associate with either the 5' or 3' end of mRNAs are in close proximity to one another during translational repression in female gametocytes and then dissociate upon release of repression in zygotes. This observation is cross-validated for several protein colocalizations in female gametocytes via ultrastructure expansion microscopy and structured illumination microscopy. Moreover, DOZI exchanges its interaction from NOT1-G in female gametocytes to the canonical NOT1 in zygotes, providing a model for a trigger for the release of mRNAs from DOZI. Finally, unenriched phosphoproteomics revealed the modification of key translational control proteins in the zygote. Together, these data provide a model for the essential translational control mechanisms used by malaria parasites to promote their efficient transmission from their mammalian host to their mosquito vector.
Collapse
Affiliation(s)
- Kelly T. Rios
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
- Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, PA, 16802
| | - James P. McGee
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
- Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, PA, 16802
| | - Aswathy Sebastian
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802
| | | | - Marina Feric
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
| | - Sabrina Absalon
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202
| | | | - Scott E. Lindner
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
- Huck Center for Malaria Research, Pennsylvania State University, University Park, PA, 16802
- Center for Eukaryotic Gene Regulation, Pennsylvania State University, University Park, PA, 16802
| |
Collapse
|
15
|
Olajiga OM, Jameson SB, Carter BH, Wesson DM, Mitzel D, Londono-Renteria B. Artificial Feeding Systems for Vector-Borne Disease Studies. BIOLOGY 2024; 13:188. [PMID: 38534457 DOI: 10.3390/biology13030188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024]
Abstract
This review examines the advancements and methodologies of artificial feeding systems for the study of vector-borne diseases, offering a critical assessment of their development, advantages, and limitations relative to traditional live host models. It underscores the ethical considerations and practical benefits of such systems, including minimizing the use of live animals and enhancing experimental consistency. Various artificial feeding techniques are detailed, including membrane feeding, capillary feeding, and the utilization of engineered biocompatible materials, with their respective applications, efficacy, and the challenges encountered with their use also being outlined. This review also forecasts the integration of cutting-edge technologies like biomimicry, microfluidics, nanotechnology, and artificial intelligence to refine and expand the capabilities of artificial feeding systems. These innovations aim to more accurately simulate natural feeding conditions, thereby improving the reliability of studies on the transmission dynamics of vector-borne diseases. This comprehensive review serves as a foundational reference for researchers in the field, proposing a forward-looking perspective on the potential of artificial feeding systems to revolutionize vector-borne disease research.
Collapse
Affiliation(s)
- Olayinka M Olajiga
- Department of Tropical Medicine and Infectious Disease, Tulane University, New Orleans, LA 70112, USA
| | - Samuel B Jameson
- Department of Tropical Medicine and Infectious Disease, Tulane University, New Orleans, LA 70112, USA
| | - Brendan H Carter
- Department of Tropical Medicine and Infectious Disease, Tulane University, New Orleans, LA 70112, USA
| | - Dawn M Wesson
- Department of Tropical Medicine and Infectious Disease, Tulane University, New Orleans, LA 70112, USA
| | - Dana Mitzel
- Animal Diseases Research Unit, National Bio- and Agro-Defense Facility, United States Department of Agriculture, Agricultural Research Service, Manhattan, KS 66506, USA
| | - Berlin Londono-Renteria
- Department of Tropical Medicine and Infectious Disease, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
16
|
N'Guessan R, Camara S, Rowland M, Ahoua Alou LP, Wolie RZ, Zoh MG, N'Guessan B, Tia IZ, Oumbouke WA, Thomas MB, Koffi AA. Attractive targeted sugar bait: the pyrrole insecticide chlorfenapyr and the anti-malarial pharmaceutical artemether-lumefantrine arrest Plasmodium falciparum development inside wild pyrethroid-resistant Anopheles gambiae s.s. mosquitoes. Malar J 2023; 22:344. [PMID: 37946208 PMCID: PMC10636997 DOI: 10.1186/s12936-023-04758-1] [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: 04/20/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Attractive targeted sugar bait (ATSB) is a novel approach to vector control, offering an alternative mode of insecticide delivery via the insect alimentary canal, with potential to deliver a variety of compounds new to medical entomology and malaria control. Its potential to control mosquitoes was recently demonstrated in major field trials in Africa. The pyrrole chlorfenapyr is an insecticide new to malaria vector control, and through its unique mode of action-disruption of ATP mediated energy transfer in mitochondria-it may have direct action on energy transfer in the flight muscle cells of mosquitoes. It may also have potential to disrupt mitochondrial function in malarial parasites co-existing within the infected mosquito. However, little is known about the impact of such compounds on vector competence in mosquitoes responsible for malaria transmission. METHODS In this study, ATSBs containing chlorfenapyr insecticide and, as a positive control, the anti-malarial drugs artemether/lumefantrine (A/L) were compared for their effect on Plasmodium falciparum development in wild pyrethroid-resistant Anopheles gambiae sensu stricto (s.s.) and for their capacity to reduce vector competence. Female mosquitoes were exposed to ATSB containing either sublethal dose of chlorfenapyr (CFP: 0.025%) or concentrations of A/L ranging from 0.4/2.4 mg/ml to 2.4/14.4 mg/ml, either shortly before or after taking infective blood meals. The impact of their component compounds on the prevalence and intensity of P. falciparum infection were compared between treatments. RESULTS Both the prevalence and intensity of infection were significantly reduced in mosquitoes exposed to either A/L or chlorfenapyr, compared to unexposed negative control mosquitoes. The A/L dose (2.4/14.4 mg/ml) totally erased P. falciparum parasites: 0% prevalence of infection in female mosquitoes exposed compared to 62% of infection in negative controls (df = 1, χ2 = 31.23 p < 0.001). The dose of chlorfenapyr (0.025%) that killed < 20% females in ATSB showed a reduction in oocyte density of 95% per midgut (0.18/3.43 per midgut). CONCLUSION These results are evidence that chlorfenapyr, in addition to its direct killing effect on the vector, has the capacity to block Plasmodium transmission by interfering with oocyte development inside pyrethroid-resistant mosquitoes, and through this dual action may potentiate its impact under field conditions.
Collapse
Affiliation(s)
- Raphael N'Guessan
- Institut Pierre Richet (IPR), Institut National de Santé Publique (INSP), Bouaké, Côte d'Ivoire. Raphael.N'
- Vector Control Product Evaluation Centre (VCPEC)-Institut Pierre Richet (VCPEC-IPR)/INSP, Bouaké, Côte d'Ivoire. Raphael.N'
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, UK. Raphael.N'
| | - Soromane Camara
- Institut Pierre Richet (IPR), Institut National de Santé Publique (INSP), Bouaké, Côte d'Ivoire.
- Vector Control Product Evaluation Centre (VCPEC)-Institut Pierre Richet (VCPEC-IPR)/INSP, Bouaké, Côte d'Ivoire.
| | - Mark Rowland
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, UK
| | - Ludovic P Ahoua Alou
- Institut Pierre Richet (IPR), Institut National de Santé Publique (INSP), Bouaké, Côte d'Ivoire
- Vector Control Product Evaluation Centre (VCPEC)-Institut Pierre Richet (VCPEC-IPR)/INSP, Bouaké, Côte d'Ivoire
| | - Rosine Z Wolie
- Vector Control Product Evaluation Centre (VCPEC)-Institut Pierre Richet (VCPEC-IPR)/INSP, Bouaké, Côte d'Ivoire
- Université Nangui Abrogoua, UFR Des Sciences de la Nature, Abidjan, Côte d'Ivoire
| | - Marius G Zoh
- Vector Control Product Evaluation Centre (VCPEC)-Institut Pierre Richet (VCPEC-IPR)/INSP, Bouaké, Côte d'Ivoire
| | - Brou N'Guessan
- Vector Control Product Evaluation Centre (VCPEC)-Institut Pierre Richet (VCPEC-IPR)/INSP, Bouaké, Côte d'Ivoire
| | - Innocent Z Tia
- Vector Control Product Evaluation Centre (VCPEC)-Institut Pierre Richet (VCPEC-IPR)/INSP, Bouaké, Côte d'Ivoire
| | - Welbeck A Oumbouke
- Vector Control Product Evaluation Centre (VCPEC)-Institut Pierre Richet (VCPEC-IPR)/INSP, Bouaké, Côte d'Ivoire
- Innovative Vector Control Consortium, IVCC, Liverpool, UK
| | - Matthew B Thomas
- Department of Entomology & Nematology, The University of Florida, Gainesville, FL, USA
| | - Alphonsine A Koffi
- Institut Pierre Richet (IPR), Institut National de Santé Publique (INSP), Bouaké, Côte d'Ivoire
- Vector Control Product Evaluation Centre (VCPEC)-Institut Pierre Richet (VCPEC-IPR)/INSP, Bouaké, Côte d'Ivoire
| |
Collapse
|
17
|
Jiang Y, Gao H, Wang L, Hu W, Wang G, Wang S. Quorum sensing-activated phenylalanine metabolism drives OMV biogenesis to enhance mosquito commensal colonization resistance to Plasmodium. Cell Host Microbe 2023; 31:1655-1667.e6. [PMID: 37738984 DOI: 10.1016/j.chom.2023.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/24/2023] [Accepted: 08/27/2023] [Indexed: 09/24/2023]
Abstract
Gut microbiota and its symbiotic relationship with the host are crucial for preventing pathogen infection. However, little is known about the mechanisms that drive commensal colonization. Serratia bacteria, commonly found in Anopheles mosquitoes, potentially mediate mosquito resistance to Plasmodium. Using S. ureilytica Su_YN1 as a model, we show that a quorum sensing (QS) circuit is crucial for stable colonization. After blood ingestion, the QS synthase SueI generates the signaling molecule N-hexanoyl-L-homoserine lactone (C6-HSL). Once C6-HSL binds to the QS receptor SueR, repression of the phenylalanine-to-acetyl-coenzyme A (CoA) conversion pathway is lifted. This pathway regulates outer membrane vesicle (OMV) biogenesis and promotes Serratia biofilm-like aggregate formation, facilitating gut adaptation and colonization. Notably, exposing Serratia Su_YN1-carrying Anopheles mosquitoes to C6-HSL increases Serratia gut colonization and enhances Plasmodium transmission-blocking efficacy. These findings provide insights into OMV biogenesis and commensal gut colonization and identify a powerful strategy for enhancing commensal resistance to pathogens.
Collapse
Affiliation(s)
- Yongmao Jiang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Han Gao
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Lihua Wang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Wenqian Hu
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Guandong Wang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Sibao Wang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
18
|
Kweyamba PA, Hofer LM, Kibondo UA, Mwanga RY, Sayi RM, Matwewe F, Austin JW, Stutz S, Moore SJ, Müller P, Tambwe MM. Sub-lethal exposure to chlorfenapyr reduces the probability of developing Plasmodium falciparum parasites in surviving Anopheles mosquitoes. Parasit Vectors 2023; 16:342. [PMID: 37789458 PMCID: PMC10546750 DOI: 10.1186/s13071-023-05963-2] [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: 07/05/2023] [Accepted: 09/06/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Pyrethroid resistance in the key malaria vectors threatens the success of pyrethroid-treated nets. To overcome pyrethroid resistance, Interceptor® G2 (IG2), a 'first-in-class' dual insecticidal net that combines alpha-cypermethrin with chlorfenapyr, was developed. Chlorfenapyr is a pro-insecticide, requiring bio-activation by oxidative metabolism within the insect's mitochondria, constituting a mode of action preventing cross-resistance to pyrethroids. Recent epidemiological trials conducted in Benin and Tanzania confirm IG2's public health value in areas with pyrethroid-resistant Anopheles mosquitoes. As chlorfenapyr might also interfere with the metabolic mechanism of the Plasmodium parasite, we hypothesised that chlorfenapyr may provide additional transmission-reducing effects even if a mosquito survives a sub-lethal dose. METHODS We tested the effect of chlorfenapyr netting to reduce Plasmodium falciparum transmission using a modified WHO tunnel test with a dose yielding sub-lethal effects. Pyrethroid-resistant Anopheles gambiae s.s. with L1014F and L1014S knockdown resistance alleles and expression levels of pyrethroid metabolisers CYP6P3, CYP6M2, CYP4G16 and CYP6P1 confirmed by quantitative reverse transcription polymerase chain reaction (RT-qPCR) prior to conducting experiments were exposed to untreated netting and netting treated with 200 mg/m3 chlorfenapyr for 8 h overnight and then fed on gametocytemic blood meals from naturally infected individuals. Prevalence and intensity of oocysts and sporozoites were determined on day 8 and day 16 after feeding. RESULTS Both prevalence and intensity of P. falciparum infection in the surviving mosquitoes were substantially reduced in the chlorfenapyr-exposed mosquitoes compared to untreated nets. The odds ratios in the prevalence of oocysts and sporozoites were 0.33 (95% confidence interval; 95% CI 0.23-0.46) and 0.43 (95% CI 0.25-0.73), respectively, while only the incidence rate ratio for oocysts was 0.30 (95% CI 0.22-0.41). CONCLUSION We demonstrated that sub-lethal exposure of pyrethroid-resistant mosquitoes to chlorfenapyr substantially reduces the proportion of infected mosquitoes and the intensity of the P. falciparum infection. This will likely also contribute to the reduction of malaria in communities beyond the direct killing of mosquitoes.
Collapse
Affiliation(s)
- Prisca A Kweyamba
- Vector Control Product Testing Unit (VCPTU), Environmental Health and Ecological Sciences, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania.
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123, Allschwil, Switzerland.
- University of Basel, Petersplatz 1, 4001, Basel, Switzerland.
| | - Lorenz M Hofer
- Vector Control Product Testing Unit (VCPTU), Environmental Health and Ecological Sciences, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123, Allschwil, Switzerland
| | - Ummi A Kibondo
- Vector Control Product Testing Unit (VCPTU), Environmental Health and Ecological Sciences, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Rehema Y Mwanga
- Vector Control Product Testing Unit (VCPTU), Environmental Health and Ecological Sciences, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Rajabu M Sayi
- Vector Control Product Testing Unit (VCPTU), Environmental Health and Ecological Sciences, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Fatuma Matwewe
- Vector Control Product Testing Unit (VCPTU), Environmental Health and Ecological Sciences, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - James W Austin
- Professional & Specialty Solutions, BASF Corporation, Global Development, Public Health Insecticides, Research Triangle Park, NC, 27709, USA
| | - Susanne Stutz
- Professional & Specialty Solutions, BASF SE, Public Health, 67117, Limburgerhof, Germany
| | - Sarah J Moore
- Vector Control Product Testing Unit (VCPTU), Environmental Health and Ecological Sciences, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123, Allschwil, Switzerland
- University of Basel, Petersplatz 1, 4001, Basel, Switzerland
- The Nelson Mandela African Institution of Science and Technology (NM-AIST), Tengeru, P.O. Box 447, Arusha, Tanzania
| | - Pie Müller
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123, Allschwil, Switzerland
- University of Basel, Petersplatz 1, 4001, Basel, Switzerland
| | - Mgeni M Tambwe
- Vector Control Product Testing Unit (VCPTU), Environmental Health and Ecological Sciences, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| |
Collapse
|
19
|
Siqueira-Neto JL, Wicht KJ, Chibale K, Burrows JN, Fidock DA, Winzeler EA. Antimalarial drug discovery: progress and approaches. Nat Rev Drug Discov 2023; 22:807-826. [PMID: 37652975 PMCID: PMC10543600 DOI: 10.1038/s41573-023-00772-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 09/02/2023]
Abstract
Recent antimalarial drug discovery has been a race to produce new medicines that overcome emerging drug resistance, whilst considering safety and improving dosing convenience. Discovery efforts have yielded a variety of new molecules, many with novel modes of action, and the most advanced are in late-stage clinical development. These discoveries have led to a deeper understanding of how antimalarial drugs act, the identification of a new generation of drug targets, and multiple structure-based chemistry initiatives. The limited pool of funding means it is vital to prioritize new drug candidates. They should exhibit high potency, a low propensity for resistance, a pharmacokinetic profile that favours infrequent dosing, low cost, preclinical results that demonstrate safety and tolerability in women and infants, and preferably the ability to block Plasmodium transmission to Anopheles mosquito vectors. In this Review, we describe the approaches that have been successful, progress in preclinical and clinical development, and existing challenges. We illustrate how antimalarial drug discovery can serve as a model for drug discovery in diseases of poverty.
Collapse
Affiliation(s)
| | - Kathryn J Wicht
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | - Kelly Chibale
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | | | - David A Fidock
- Department of Microbiology and Immunology and Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | |
Collapse
|
20
|
Wang L, Sanon A, Khoiriyah Z, Verwimp S, Abdelnabi R, Delang L. Tarsal exposure to atovaquone inhibits chikungunya virus transmission by Aedes aegypti mosquitoes, but not the transmission of Zika virus. Antiviral Res 2023; 217:105694. [PMID: 37532005 DOI: 10.1016/j.antiviral.2023.105694] [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: 05/09/2023] [Revised: 07/10/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
The antimalarial drug atovaquone was recently reported to inhibit the in vitro replication of different arboviruses, including chikungunya virus (CHIKV) and Zika virus (ZIKV). Furthermore, atovaquone was shown to block Plasmodium parasite transmission by Anopheles mosquitoes when the mosquitoes were exposed to low concentrations on treated surfaces (i.e. tarsal exposure). Therefore, we evaluated the anti-CHIKV and -ZIKV effects of atovaquone via tarsal exposure in Aedes aegypti mosquitoes. We first confirmed that atovaquone exerted a dose-dependent antiviral effect on CHIKV and ZIKV replication in mosquito-derived cells. The modest antiviral effect could be rescued by adding exogenous uridine. Next, we assessed the effect of tarsal exposure to atovaquone on the fitness of Ae. aegypti. Concentrations up to 100 μmol/m2 did not affect the fecundity and egg-hatching rate. No significant effect on mosquito survival was observed when mosquitoes were exposed to concentrations up to 25 μmol/m2. To evaluate the antiviral effect of atovaquone against CHIKV, we exposed female mosquitoes to 100 μmol/m2 atovaquone for 1h, after which the mosquitoes were immediately infected with CHIKV or ZIKV via bloodmeal. Atovaquone did not significantly reduce ZIKV or CHIKV infection in Ae. aegypti, but successfully blocked the transmission of CHIKV in saliva. Tarsal exposure to antiviral drugs could therefore be a potential new strategy to reduce virus transmission by mosquitoes.
Collapse
Affiliation(s)
- Lanjiao Wang
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Aboubakar Sanon
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium; Laboratoire d'Entomologie Fondamentale et Appliquée, Université Joseph Ki-Zerbo, Burkina Faso
| | - Zakiyatul Khoiriyah
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium; Laboratory of Virology, Wageningen University and Research, 6708 PB, Wageningen, the Netherlands
| | - Sam Verwimp
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Rana Abdelnabi
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Leen Delang
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium.
| |
Collapse
|
21
|
Huang W, Rodrigues J, Bilgo E, Tormo JR, Challenger JD, De Cozar-Gallardo C, Pérez-Victoria I, Reyes F, Castañeda-Casado P, Gnambani EJ, Hien DFDS, Konkobo M, Urones B, Coppens I, Mendoza-Losana A, Ballell L, Diabate A, Churcher TS, Jacobs-Lorena M. Delftia tsuruhatensis TC1 symbiont suppresses malaria transmission by anopheline mosquitoes. Science 2023; 381:533-540. [PMID: 37535741 DOI: 10.1126/science.adf8141] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 06/22/2023] [Indexed: 08/05/2023]
Abstract
Malaria control demands the development of a wide range of complementary strategies. We describe the properties of a naturally occurring, non-genetically modified symbiotic bacterium, Delftia tsuruhatensis TC1, which was isolated from mosquitoes incapable of sustaining the development of Plasmodium falciparum parasites. D. tsuruhatensis TC1 inhibits early stages of Plasmodium development and subsequent transmission by the Anopheles mosquito through secretion of a small-molecule inhibitor. We have identified this inhibitor to be the hydrophobic molecule harmane. We also found that, on mosquito contact, harmane penetrates the cuticle, inhibiting Plasmodium development. D. tsuruhatensis TC1 stably populates the mosquito gut, does not impose a fitness cost on the mosquito, and inhibits Plasmodium development for the mosquito's life. Contained field studies in Burkina Faso and modeling showed that D. tsuruhatensis TC1 has the potential to complement mosquito-targeted malaria transmission control.
Collapse
Affiliation(s)
- Wei Huang
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Etienne Bilgo
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso BP: 545, Burkina Faso
| | | | - Joseph D Challenger
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London W2 1PG, UK
| | | | | | | | - Pablo Castañeda-Casado
- Drug Metabolism and Pharmacokinetics (DMPK) Discovery, In Vitro/In Vivo Translation (IVIVT), GSK, 28760 Tres Cantos, Madrid, Spain
| | | | | | - Maurice Konkobo
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso BP: 545, Burkina Faso
| | - Beatriz Urones
- Global Health Medicines R&D, GSK, Tres Cantos, 28760 Madrid, Spain
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Lluís Ballell
- Global Health Medicines R&D, GSK, Tres Cantos, 28760 Madrid, Spain
| | - Abdoulaye Diabate
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso BP: 545, Burkina Faso
| | - Thomas S Churcher
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London W2 1PG, UK
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
22
|
B. Henry N, Soulama I, S. Sermé S, Bolscher JM, T. G. Huijs T, S. Coulibaly A, Sombié S, Ouédraogo N, Diarra A, Zongo S, Guelbéogo WM, Nébié I, Sirima SB, Tiono AB, Pietro A, Collins KA, Dechering KJ, Bousema T. Assessment of the transmission blocking activity of antimalarial compounds by membrane feeding assays using natural Plasmodium falciparum gametocyte isolates from West-Africa. PLoS One 2023; 18:e0284751. [PMID: 37494413 PMCID: PMC10370769 DOI: 10.1371/journal.pone.0284751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 04/07/2023] [Indexed: 07/28/2023] Open
Abstract
Antimalarial drugs that can block the transmission of Plasmodium gametocytes to mosquito vectors would be highly beneficial for malaria elimination efforts. Identifying transmission-blocking drugs currently relies on evaluation of their activity against gametocyte-producing laboratory parasite strains and would benefit from a testing pipeline with genetically diverse field isolates. The aims of this study were to develop a pipeline to test drugs against P. falciparum gametocyte field isolates and to evaluate the transmission-blocking activity of a set of novel compounds. Two assays were designed so they could identify both the overall transmission-blocking activity of a number of marketed and experimental drugs by direct membrane feeding assays (DMFA), and then also discriminate between those that are active against the gametocytes (gametocyte killing or sterilizing) or those that block development in the mosquito (sporontocidal). These DMFA assays used venous blood samples from naturally infected Plasmodium falciparum gametocyte carriers and locally reared Anopheles gambiae s.s. mosquitoes. Overall transmission-blocking activity was assessed following a 24 hour incubation of compound with gametocyte infected blood (TB-DMFA). Sporontocidal activity was evaluated following addition of compound directly prior to feeding, without incubation (SPORO-DMFA); Gametocyte viability was retained during 24-hour incubation at 37°C when gametocyte infected red blood cells were reconstituted in RPMI/serum. Methylene-blue, MMV693183, DDD107498, atovaquone and P218 showed potent transmission-blocking activity in the TB-DMFA, and both atovaquone and the novel antifolate P218 were potent inhibitors of sporogonic development in the SPORO-DMA. This work establishes a pipeline for the integral use of field isolates to assess the transmission-blocking capacity of antimalarial drugs to block transmission that should be validated in future studies.
Collapse
Affiliation(s)
- Noëlie B. Henry
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Issiaka Soulama
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
- Institut de Recherche en Sciences de la Santé (IRSS)/CNRST, Ouagadougou, Burkina Faso
| | - Samuel S. Sermé
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | | | | | - Aboubacar S. Coulibaly
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Salif Sombié
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Nicolas Ouédraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Amidou Diarra
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Soumanaba Zongo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Wamdaogo M. Guelbéogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Issa Nébié
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso
| | | | - Alfred B. Tiono
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Alano Pietro
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Katharine A. Collins
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherland
| | | | - Teun Bousema
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherland
| |
Collapse
|
23
|
Espino-Sanchez T, Wienkers H, Marvin R, Nalder SA, García-Guerrero A, VanNatta P, Jami-Alahmadi Y, Mixon Blackwell A, Whitby F, Wohlschlegel J, Kieber-Emmons M, Hill C, A. Sigala P. Direct tests of cytochrome c and c1 functions in the electron transport chain of malaria parasites. Proc Natl Acad Sci U S A 2023; 120:e2301047120. [PMID: 37126705 PMCID: PMC10175771 DOI: 10.1073/pnas.2301047120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/30/2023] [Indexed: 05/03/2023] Open
Abstract
The mitochondrial electron transport chain (ETC) of Plasmodium malaria parasites is a major antimalarial drug target, but critical cytochrome (cyt) functions remain unstudied and enigmatic. Parasites express two distinct cyt c homologs (c and c-2) with unusually sparse sequence identity and uncertain fitness contributions. P. falciparum cyt c-2 is the most divergent eukaryotic cyt c homolog currently known and has sequence features predicted to be incompatible with canonical ETC function. We tagged both cyt c homologs and the related cyt c1 for inducible knockdown. Translational repression of cyt c and cyt c1 was lethal to parasites, which died from ETC dysfunction and impaired ubiquinone recycling. In contrast, cyt c-2 knockdown or knockout had little impact on blood-stage growth, indicating that parasites rely fully on the more conserved cyt c for ETC function. Biochemical and structural studies revealed that both cyt c and c-2 are hemylated by holocytochrome c synthase, but UV-vis absorbance and EPR spectra strongly suggest that cyt c-2 has an unusually open active site in which heme is stably coordinated by only a single axial amino acid ligand and can bind exogenous small molecules. These studies provide a direct dissection of cytochrome functions in the ETC of malaria parasites and identify a highly divergent Plasmodium cytochrome c with molecular adaptations that defy a conserved role in eukaryotic evolution.
Collapse
Affiliation(s)
| | - Henry Wienkers
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Rebecca G. Marvin
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Shai-anne Nalder
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | | | - Peter E. VanNatta
- Department of Chemistry, University of Utah, Salt Lake City, UT84112
| | | | - Amanda Mixon Blackwell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Frank G. Whitby
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | | | | | - Christopher P. Hill
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| |
Collapse
|
24
|
Yoshinaga M, Niu G, Yoshinaga-Sakurai K, Nadar VS, Wang X, Rosen BP, Li J. Arsinothricin Inhibits Plasmodium falciparum Proliferation in Blood and Blocks Parasite Transmission to Mosquitoes. Microorganisms 2023; 11:1195. [PMID: 37317169 PMCID: PMC10222646 DOI: 10.3390/microorganisms11051195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023] Open
Abstract
Malaria, caused by Plasmodium protozoal parasites, remains a leading cause of morbidity and mortality. The Plasmodium parasite has a complex life cycle, with asexual and sexual forms in humans and Anopheles mosquitoes. Most antimalarials target only the symptomatic asexual blood stage. However, to ensure malaria eradication, new drugs with efficacy at multiple stages of the life cycle are necessary. We previously demonstrated that arsinothricin (AST), a newly discovered organoarsenical natural product, is a potent broad-spectrum antibiotic that inhibits the growth of various prokaryotic pathogens. Here, we report that AST is an effective multi-stage antimalarial. AST is a nonproteinogenic amino acid analog of glutamate that inhibits prokaryotic glutamine synthetase (GS). Phylogenetic analysis shows that Plasmodium GS, which is expressed throughout all stages of the parasite life cycle, is more closely related to prokaryotic GS than eukaryotic GS. AST potently inhibits Plasmodium GS, while it is less effective on human GS. Notably, AST effectively inhibits both Plasmodium erythrocytic proliferation and parasite transmission to mosquitoes. In contrast, AST is relatively nontoxic to a number of human cell lines, suggesting that AST is selective against malaria pathogens, with little negative effect on the human host. We propose that AST is a promising lead compound for developing a new class of multi-stage antimalarials.
Collapse
Affiliation(s)
- Masafumi Yoshinaga
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St., Miami, FL 33199, USA
| | - Guodong Niu
- Department of Biological Sciences, College of Arts, Sciences & Education, Florida International University, 11200 SW 8th St., Miami, FL 33199, USA
| | - Kunie Yoshinaga-Sakurai
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St., Miami, FL 33199, USA
| | - Venkadesh S. Nadar
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St., Miami, FL 33199, USA
| | - Xiaohong Wang
- Department of Biological Sciences, College of Arts, Sciences & Education, Florida International University, 11200 SW 8th St., Miami, FL 33199, USA
| | - Barry P. Rosen
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th St., Miami, FL 33199, USA
| | - Jun Li
- Department of Biological Sciences, College of Arts, Sciences & Education, Florida International University, 11200 SW 8th St., Miami, FL 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
25
|
Carson J, Erriah B, Herodotou S, Shtukenberg AG, Smith L, Ryazanskaya S, Ward MD, Kahr B, Lees RS. Overcoming insecticide resistance in Anopheles mosquitoes by using faster-acting solid forms of deltamethrin. Malar J 2023; 22:129. [PMID: 37081532 PMCID: PMC10120210 DOI: 10.1186/s12936-023-04554-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/05/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND Controlling malaria-transmitting Anopheles mosquitoes with pyrethroid insecticides is becoming increasingly challenging because of widespread resistance amongst vector populations. The development of new insecticides and insecticidal formulations is time consuming and costly, however. A more active crystalline form of deltamethrin, prepared by heating the commercial crystalline form, previously was reported to be 12-times faster acting against susceptible North American Anopheles quadrimaculatus mosquitoes. Herein the potential for heat-activated deltamethrin dispersed on chalk to overcome various resistance mechanisms amongst five West African Anopheles strains is investigated, and its long-term sustained lethality evaluated. METHODS The more active deltamethrin form was generated in a commercial dust containing deltamethrin by heating the material as purchased. Tarsal contact bioassays were conducted to investigate its efficacy, potency, and speed of action against resistant Anopheles populations compared to the commercially available form of deltamethrin dust. RESULTS In all cases, D-Fense Dust heated to generate the more active form of deltamethrin was substantially more effective than the commercially available formulation. 100% of both Banfora M and Kisumu populations were knocked down 10 min post-exposure with no recovery afterwards. Gaoua-ara and Tiefora strains exhibited 100% knockdown within 15 min, and the VK7 2014 strain exhibited 100% knockdown within 20 min. In all cases, 100% mortality was observed 24 h post-exposure. Conversely, the commercial formulation (unheated) resulted in less than 4% mortality amongst VK7 2014, Banfora, and Gaoua-ara populations by 24 h, and Tiefora and Kisumu mosquitoes experienced 14 and 47% mortality by 24 h, respectively. The heat-activated dust maintained comparable efficacy 13 months after heating. CONCLUSIONS The heat-activated form of commercial deltamethrin D-Fense Dust outperformed the material as purchased, dramatically increasing efficacy against all tested pyrethroid-resistant strains. This increase in lethality was retained for 13 months of storage under ambient conditions in the laboratory. Higher energy forms of commonly used insecticides may be employed to overcome various resistance mechanisms seen in African Anopheles mosquitoes through more rapid uptake of insecticide molecules from their respective solid surfaces. That is, resistant mosquitoes can be killed with an insecticide to which they are resistant without altering the molecular composition of the insecticide.
Collapse
Affiliation(s)
- Jessica Carson
- Vector Biology Department, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Bryan Erriah
- Department of Chemistry and Molecular Design Institute, New York University, 29 Washington Place, New York, 10003, NY, USA
| | - Stephania Herodotou
- Liverpool School of Tropical Medicine, Innovative Vector Control Consortium, Pembroke Place, Liverpool, L3 5QA, UK
| | - Alexander G Shtukenberg
- Department of Chemistry and Molecular Design Institute, New York University, 29 Washington Place, New York, 10003, NY, USA
| | - Leilani Smith
- Department of Chemistry and Molecular Design Institute, New York University, 29 Washington Place, New York, 10003, NY, USA
| | - Svetlana Ryazanskaya
- Liverpool School of Tropical Medicine, Innovative Vector Control Consortium, Pembroke Place, Liverpool, L3 5QA, UK
| | - Michael D Ward
- Department of Chemistry and Molecular Design Institute, New York University, 29 Washington Place, New York, 10003, NY, USA
| | - Bart Kahr
- Department of Chemistry and Molecular Design Institute, New York University, 29 Washington Place, New York, 10003, NY, USA
| | - Rosemary Susan Lees
- Vector Biology Department, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| |
Collapse
|
26
|
Calit J, Araújo JE, Deng B, Miura K, Gaitán XA, Araújo MDS, Medeiros JF, Long CA, Simeonov A, Eastman RT, Bargieri DY. Novel Transmission-Blocking Antimalarials Identified by High-Throughput Screening of Plasmodium berghei Ookluc. Antimicrob Agents Chemother 2023; 67:e0146522. [PMID: 36856421 PMCID: PMC10112123 DOI: 10.1128/aac.01465-22] [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: 10/31/2022] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Safe and effective malaria transmission-blocking chemotherapeutics would allow a community-level approach to malaria control and eradication efforts by targeting the mosquito sexual stage of the parasite life cycle. However, only a single drug, primaquine, is currently approved for use in reducing transmission, and drug toxicity limits its widespread implementation. To address this limitation in antimalarial chemotherapeutics, we used a recently developed transgenic Plasmodium berghei line, Ookluc, to perform a series of high-throughput in vitro screens for compounds that inhibit parasite fertilization, the initial step of parasite development within the mosquito. Screens of antimalarial compounds, approved drug collections, and drug-like molecule libraries identified 185 compounds that inhibit parasite maturation to the zygote form. Seven compounds were further characterized to block gametocyte activation or to be cytotoxic to formed zygotes. These were further validated in mosquito membrane-feeding assays using Plasmodium falciparum and P. vivax. This work demonstrates that high-throughput screens using the Ookluc line can identify compounds that are active against the two most relevant human Plasmodium species and provides a list of compounds that can be explored for the development of new antimalarials to block transmission.
Collapse
Affiliation(s)
- Juliana Calit
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Jessica E. Araújo
- Plataforma de Produção e Infecção de Vetores da Malária-PIVEM, Laboratório de Entomologia, Fundação Oswaldo Cruz-Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia/Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Bingbing Deng
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Xiomara A. Gaitán
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Maisa da Silva Araújo
- Plataforma de Produção e Infecção de Vetores da Malária-PIVEM, Laboratório de Entomologia, Fundação Oswaldo Cruz-Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Jansen F. Medeiros
- Plataforma de Produção e Infecção de Vetores da Malária-PIVEM, Laboratório de Entomologia, Fundação Oswaldo Cruz-Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia/Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Richard T. Eastman
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Daniel Y. Bargieri
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
27
|
Perugini E, Guelbeogo WM, Guglielmo F, Poggi C, Gabrieli E, Ranson H, Della Torre A, Pombi M. The interplay between malaria vectors and human activity accounts for high residual malaria transmission in a Burkina Faso village with universal ITN coverage. Parasit Vectors 2023; 16:101. [PMID: 36922855 PMCID: PMC10015820 DOI: 10.1186/s13071-023-05710-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/22/2023] [Indexed: 03/17/2023] Open
Abstract
BACKGROUND Mosquito and human behaviour interaction is a key determinant of the maximum level of protection against malaria that can be provided by insecticide-treated nets (ITNs). Nevertheless, scant literature focuses on this interaction, overlooking a fundamental factor for efficient malaria control. This study aims to estimate malaria transmission risk in a Burkina Faso village by integrating vector biting rhythms with some key information about human habits. METHODS Indoor/outdoor human landing catches were conducted for 16 h (16:00-08:00) during 8 nights (September 2020) in Goden village. A survey about net usage and sleeping patterns was submitted to half the households (October-December 2020). A subsample of collected specimens of Anopheles gambiae sensu lato was molecularly processed for species identification, Plasmodium detection from heads-thoraxes and L1014F pyrethroid-resistance allele genotyping. Hourly mosquito abundance was statistically assessed by GLM/GAM, and the entomological inoculation rate (EIR) was corrected for the actual ITN usage retrieved from the questionnaire. RESULTS Malaria transmission was mainly driven by Anopheles coluzzii (68.7%) followed by A. arabiensis (26.2%). The overall sporozoite rate was 2% with L1014F estimated frequency of 0.68 (N = 1070 out of 15,201 A. gambiae s.l. collected). No major shift in mosquito biting rhythms in response to ITN or differences between indoor and outdoor catches were detected. Impressive high biting pressure (mean 30.3 mosquitoes/person/hour) was exerted from 20:00 to 06:00 with a peak at 4:00. Human survey revealed that nearly all inhabitants were awake before 20:00 and after 7:00 and at least 8.7% had no access to bednets. Adjusting for anthropological data, the EIR dropped from 6.7 to 1.2 infective bites/person/16 h. In a scenario of full net coverage and accounting only for the human sleeping patterns, the daily malaria transmission risk not targetable by ITNs was 0.69 infective bites. CONCLUSIONS The high mosquito densities and interplay between human/vector activities means that an estimated 10% of residual malaria transmission cannot be prevented by ITNs in the village. Locally tailored studies, like the current one, are essential to explore the heterogeneity of human exposure to infective bites and, consequently, to instruct the adoption of new vector control tools strengthening individual and community protection.
Collapse
Affiliation(s)
- Eleonora Perugini
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Wamdaogo M Guelbeogo
- Centre National de Recherche et Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Federica Guglielmo
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Cristiana Poggi
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Eugenio Gabrieli
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Hilary Ranson
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Marco Pombi
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy.
| |
Collapse
|
28
|
Mohammed M, Dziedziech A, Sekar V, Ernest M, Alves E Silva TL, Balan B, Emami SN, Biryukova I, Friedländer MR, Jex A, Jacobs-Lorena M, Henriksson J, Vega-Rodriguez J, Ankarklev J. Single-Cell Transcriptomics To Define Plasmodium falciparum Stage Transition in the Mosquito Midgut. Microbiol Spectr 2023; 11:e0367122. [PMID: 36847501 PMCID: PMC10100735 DOI: 10.1128/spectrum.03671-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 02/05/2023] [Indexed: 03/01/2023] Open
Abstract
Malaria inflicts the highest rate of morbidity and mortality among the vector-borne diseases. The dramatic bottleneck of parasite numbers that occurs in the gut of the obligatory mosquito vector provides a promising target for novel control strategies. Using single-cell transcriptomics, we analyzed Plasmodium falciparum development in the mosquito gut, from unfertilized female gametes through the first 20 h after blood feeding, including the zygote and ookinete stages. This study revealed the temporal gene expression of the ApiAP2 family of transcription factors and of parasite stress genes in response to the harsh environment of the mosquito midgut. Further, employing structural protein prediction analyses, we found several upregulated genes predicted to encode intrinsically disordered proteins (IDPs), a category of proteins known for their importance in regulation of transcription, translation, and protein-protein interactions. IDPs are known for their antigenic properties and may serve as suitable targets for antibody- or peptide-based transmission suppression strategies. In total, this study uncovers the P. falciparum transcriptome from early to late parasite development in the mosquito midgut, inside its natural vector, which provides an important resource for future malaria transmission-blocking initiatives. IMPORTANCE The malaria parasite Plasmodium falciparum causes more than half a million deaths per year. The current treatment regimen targets the symptom-causing blood stage inside the human host. However, recent incentives in the field call for novel interventions to block parasite transmission from humans to the mosquito vector. Therefore, we need to better understand the parasite biology during its development inside the mosquito, including a deeper understanding of the expression of genes controlling parasite progression during these stages. Here, we have generated single-cell transcriptome data, covering P. falciparum's development, from gamete to ookinete inside the mosquito midgut, uncovering previously untapped parasite biology, including a repertoire of novel biomarkers to be explored in future transmission-blocking efforts. We anticipate that our study provides an important resource, which can be further explored to improve our understanding of the parasite biology as well as aid in guiding future malaria intervention strategies.
Collapse
Affiliation(s)
- Mubasher Mohammed
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Alexis Dziedziech
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Vaishnovi Sekar
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Medard Ernest
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Thiago Luiz Alves E Silva
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Balu Balan
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - S. Noushin Emami
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Inna Biryukova
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Marc R. Friedländer
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Aaron Jex
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Johan Henriksson
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Joel Vega-Rodriguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Johan Ankarklev
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- Microbial Single Cell Genomics, Department of Cell and Molecular Biology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
29
|
First person – Sabrina Yahiya. Dis Model Mech 2023. [DOI: 10.1242/dmm.050069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
ABSTRACT
First Person is a series of interviews with the first authors of a selection of papers published in Disease Models & Mechanisms, helping researchers promote themselves alongside their papers. Sabrina Yahiya is first author on ‘ A novel class of sulphonamides potently block malaria transmission by targeting a Plasmodium vacuole membrane protein’, published in DMM. Sabrina conducted the research described in this article while a PhD candidate in Prof. Jake Baum's lab at Imperial College London, UK. She is now a scientist in the Infectious Diseases & Vaccines Group at Kymab, a Sanofi company, in Cambridge, UK, and is interested in the development of therapeutics targeting infectious diseases, with a focus on blocking malaria transmission.
Collapse
|
30
|
Yahiya S, Saunders CN, Hassan S, Straschil U, Fischer OJ, Rueda-Zubiaurre A, Haase S, Vizcay-Barrena G, Famodimu MT, Jordan S, Delves MJ, Tate EW, Barnard A, Fuchter MJ, Baum J. A novel class of sulphonamides potently block malaria transmission by targeting a Plasmodium vacuole membrane protein. Dis Model Mech 2023; 16:dmm049950. [PMID: 36715290 PMCID: PMC9934914 DOI: 10.1242/dmm.049950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/13/2022] [Indexed: 01/31/2023] Open
Abstract
Phenotypic cell-based screens are critical tools for discovering candidate drugs for development, yet identification of the cellular target and mode of action of a candidate drug is often lacking. Using an imaging-based screen, we recently discovered an N-[(4-hydroxychroman-4-yl)methyl]-sulphonamide (N-4HCS) compound, DDD01035881, that blocks male gamete formation in the malaria parasite life cycle and subsequent transmission of the parasite to the mosquito with nanomolar activity. To identify the target(s) of DDD01035881, and of the N-4HCS class of compounds more broadly, we synthesised a photoactivatable derivative, probe 2. Photoaffinity labelling of probe 2 coupled with mass spectrometry identified the 16 kDa Plasmodium falciparum parasitophorous vacuole membrane protein Pfs16 as a potential parasite target. Complementary methods including cellular thermal shift assays confirmed that the parent molecule DDD01035881 stabilised Pfs16 in lysates from activated mature gametocytes. Combined with high-resolution, fluorescence and electron microscopy data, which demonstrated that parasites inhibited with N-4HCS compounds phenocopy the targeted deletion of Pfs16 in gametocytes, these data implicate Pfs16 as a likely target of DDD01035881. This finding establishes N-4HCS compounds as being flexible and effective starting candidates from which transmission-blocking antimalarials can be developed in the future.
Collapse
Affiliation(s)
- Sabrina Yahiya
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK
| | - Charlie N. Saunders
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Sarah Hassan
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Ursula Straschil
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK
| | - Oliver J. Fischer
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Ainoa Rueda-Zubiaurre
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Silvia Haase
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK
| | - Gema Vizcay-Barrena
- Centre for Ultrastructural Imaging, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Mufuliat Toyin Famodimu
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK
| | - Sarah Jordan
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK
| | - Michael J. Delves
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK
| | - Edward W. Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Anna Barnard
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Matthew J. Fuchter
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK
| |
Collapse
|
31
|
Espino-Sanchez TJ, Wienkers H, Marvin RG, Nalder SA, García-Guerrero AE, VanNatta PE, Jami-Alahmadi Y, Blackwell AM, Whitby FG, Wohlschlegel JA, Kieber-Emmons MT, Hill CP, Sigala PA. Direct Tests of Cytochrome Function in the Electron Transport Chain of Malaria Parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.525242. [PMID: 36747727 PMCID: PMC9900762 DOI: 10.1101/2023.01.23.525242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The mitochondrial electron transport chain (ETC) of Plasmodium malaria parasites is a major antimalarial drug target, but critical cytochrome functions remain unstudied and enigmatic. Parasites express two distinct cyt c homologs ( c and c -2) with unusually sparse sequence identity and uncertain fitness contributions. P. falciparum cyt c -2 is the most divergent eukaryotic cyt c homolog currently known and has sequence features predicted to be incompatible with canonical ETC function. We tagged both cyt c homologs and the related cyt c 1 for inducible knockdown. Translational repression of cyt c and cyt c 1 was lethal to parasites, which died from ETC dysfunction and impaired ubiquinone recycling. In contrast, cyt c -2 knockdown or knock-out had little impact on blood-stage growth, indicating that parasites rely fully on the more conserved cyt c for ETC function. Biochemical and structural studies revealed that both cyt c and c -2 are hemylated by holocytochrome c synthase, but UV-vis absorbance and EPR spectra strongly suggest that cyt c -2 has an unusually open active site in which heme is stably coordinated by only a single axial amino-acid ligand and can bind exogenous small molecules. These studies provide a direct dissection of cytochrome functions in the ETC of malaria parasites and identify a highly divergent Plasmodium cytochrome c with molecular adaptations that defy a conserved role in eukaryotic evolution. SIGNIFICANCE STATEMENT Mitochondria are critical organelles in eukaryotic cells that drive oxidative metabolism. The mitochondrion of Plasmodium malaria parasites is a major drug target that has many differences from human cells and remains poorly studied. One key difference from humans is that malaria parasites express two cytochrome c proteins that differ significantly from each other and play untested and uncertain roles in the mitochondrial electron transport chain (ETC). Our study revealed that one cyt c is essential for ETC function and parasite viability while the second, more divergent protein has unusual structural and biochemical properties and is not required for growth of blood-stage parasites. This work elucidates key biochemical properties and evolutionary differences in the mitochondrial ETC of malaria parasites.
Collapse
Affiliation(s)
- Tanya J. Espino-Sanchez
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Henry Wienkers
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Rebecca G. Marvin
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Shai-anne Nalder
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Aldo E. García-Guerrero
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Peter E. VanNatta
- Department of Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, University of California, Los Angeles, CA, United States
| | - Amanda Mixon Blackwell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Frank G. Whitby
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - James A. Wohlschlegel
- Department of Biological Chemistry, University of California, Los Angeles, CA, United States
| | | | - Christopher P. Hill
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
32
|
Hu B, Yan W, Jiang P, Jiang L, Yuan X, Lin J, Jiao Y, Jin Y. Switchable synthesis of natural-product-like lawsones and indenopyrazoles through regioselective ring-expansion of indantrione. Commun Chem 2023; 6:17. [PMID: 36697885 PMCID: PMC9849474 DOI: 10.1038/s42004-022-00807-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 12/29/2022] [Indexed: 01/19/2023] Open
Abstract
Lawsones and indenopyrazoles are the prevalent structural motifs and building blocks in pharmaceuticals and bioactive molecules, but their synthesis has always remained challenging as no comprehensive protocol has been outlined to date. Herein, a metal-free, ring-expansion reaction of indantrione with diazomethanes, generated in situ from the N-tosylhydrazones, has been developed for the synthesis of lawsone and indenopyrazole derivatives in acetonitrile and alcohol solvents, respectively. It provides these valuable lawsone and pyrazole skeletons in good yields and high levels of diastereoselectivity from simple and readily available starting materials. DFT calculations were used to explore the mechanism in different solutions. The synthetic application example also showed the prospects of this method for the preparation of valuable compounds.
Collapse
Affiliation(s)
- Bingwei Hu
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China
| | - Wenxin Yan
- School of Chemistry and Chemical Engineering, Key Laboratory of Theoretical Organic Chemistry and Functional Molecular, Ministry of Education, Hunan University of Science and Technology, 411201, Xiangtan, China
| | - Peiyun Jiang
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China
| | - Ling Jiang
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China
| | - Xu Yuan
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China
| | - Jun Lin
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China
| | - Yinchun Jiao
- School of Chemistry and Chemical Engineering, Key Laboratory of Theoretical Organic Chemistry and Functional Molecular, Ministry of Education, Hunan University of Science and Technology, 411201, Xiangtan, China.
| | - Yi Jin
- Key Laboratory of Medicinal for Natural Resource, Ministry of Education and Yunnan Province, School of Pharmacy, Yunnan University, 650091, Kunming, China.
| |
Collapse
|
33
|
Habtamu K, Petros B, Yan G. Plasmodium vivax: the potential obstacles it presents to malaria elimination and eradication. Trop Dis Travel Med Vaccines 2022; 8:27. [PMID: 36522671 PMCID: PMC9753897 DOI: 10.1186/s40794-022-00185-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
Initiatives to eradicate malaria have a good impact on P. falciparum malaria worldwide. P. vivax, however, still presents significant difficulties. This is due to its unique biological traits, which, in comparison to P. falciparum, pose serious challenges for malaria elimination approaches. P. vivax's numerous distinctive characteristics and its ability to live for weeks to years in liver cells in its hypnozoite form, which may elude the human immune system and blood-stage therapy and offer protection during mosquito-free seasons. Many malaria patients are not fully treated because of contraindications to primaquine use in pregnant and nursing women and are still vulnerable to P. vivax relapses, although there are medications that could radical cure P. vivax. Additionally, due to CYP2D6's highly variable genetic polymorphism, the pharmacokinetics of primaquine may be impacted. Due to their inability to metabolize PQ, some CYP2D6 polymorphism alleles can cause patients to not respond to treatment. Tafenoquine offers a radical treatment in a single dose that overcomes the potentially serious problem of poor adherence to daily primaquine. Despite this benefit, hemolysis of the early erythrocytes continues in individuals with G6PD deficiency until all susceptible cells have been eliminated. Field techniques such as microscopy or rapid diagnostic tests (RDTs) miss the large number of submicroscopic and/or asymptomatic infections brought on by reticulocyte tropism and the low parasitemia levels that accompany it. Moreover, P. vivax gametocytes grow more quickly and are much more prevalent in the bloodstream. P. vivax populations also have a great deal of genetic variation throughout their genome, which ensures evolutionary fitness and boosts adaptation potential. Furthermore, P. vivax fully develops in the mosquito faster than P. falciparum. These characteristics contribute to parasite reservoirs in the human population and facilitate faster transmission. Overall, no genuine chance of eradication is predicted in the next few years unless new tools for lowering malaria transmission are developed (i.e., malaria elimination and eradication). The challenging characteristics of P. vivax that impede the elimination and eradication of malaria are thus discussed in this article.
Collapse
Affiliation(s)
- Kassahun Habtamu
- Department of Microbial, Cellular & Molecular Biology, Addis Ababa University, Addis Ababa, Ethiopia
- Menelik II Medical & Health Science College, Addis Ababa, Ethiopia
| | - Beyene Petros
- Department of Microbial, Cellular & Molecular Biology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Guiyun Yan
- Program in Public Health, University of California at Irvine, Irvine, CA 92697 USA
| |
Collapse
|
34
|
Saison N, Franetich JF, Pinilla YT, Hoffmann A, Boussougou-Sambe ST, Ngossanga B, Tefit M, Ashraf K, Amanzougaghene N, Tajeri S, Adegnika AA, Mazier D, Borrmann S. Rapid and Specific Action of Methylene Blue against Plasmodium Transmission Stages. Pharmaceutics 2022; 14:pharmaceutics14122794. [PMID: 36559287 PMCID: PMC9786052 DOI: 10.3390/pharmaceutics14122794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Methylene blue (MB) is the oldest synthetic anti-infective. Its high potency against asexual and sexual stages of malaria parasites is well documented. This study aimed to investigate possible additional activities of MB in interfering with parasite transmission and determine target stages in Anopheles vectors and humans. MB's transmission-blocking activity was first evaluated by an ex vivo direct membrane feeding assay (DMFA) using Plasmodium falciparum field isolates. To investigate anti-mosquito stage activity, Plasmodium berghei-infected Anopheles stephensi mosquitoes were fed a second blood meal on mice that had been treated with methylene blue, 3, 6- and 15-days after the initial infectious blood meal. Anti-sporozoite and liver stage activities were evaluated in vitro and in vivo via sporozoite invasion and liver stage development assays, respectively. MB exhibited a robust inhibition of P. falciparum transmission in An. gambiae, even when added shortly before the DMFA but only a moderate effect against P. berghei oocyst development. Exposure of mature P. berghei and P. falciparum sporozoites to MB blocked hepatocyte invasion, yet P. berghei liver stage development was unaffected by MB. Our results indicate previously underappreciated rapid specific activities of methylene blue against Plasmodium transmission stages, preventing the establishment of both mosquito midgut and liver infections as the first essential steps in both hosts.
Collapse
Affiliation(s)
- Nathanaël Saison
- Centre de Recherches Médicales de Lambaréné, Lambaréné 1437, Gabon
- Institute for Tropical Medicine, University of Tübingen, 72074 Tübingen, Germany
| | - Jean-François Franetich
- Centre d’Immunologie et des Maladies Infectieuses, Inserm, CNRS, Sorbonne Université, U1135, ERL8255, CIMI-Paris, F-75013 Paris, France
| | - Yudi T. Pinilla
- Institute for Tropical Medicine, University of Tübingen, 72074 Tübingen, Germany
| | - Anton Hoffmann
- Centre de Recherches Médicales de Lambaréné, Lambaréné 1437, Gabon
- Institute for Tropical Medicine, University of Tübingen, 72074 Tübingen, Germany
| | - Stravensky T. Boussougou-Sambe
- Centre de Recherches Médicales de Lambaréné, Lambaréné 1437, Gabon
- Institute for Tropical Medicine, University of Tübingen, 72074 Tübingen, Germany
| | | | - Maurel Tefit
- Centre d’Immunologie et des Maladies Infectieuses, Inserm, CNRS, Sorbonne Université, U1135, ERL8255, CIMI-Paris, F-75013 Paris, France
| | - Kutub Ashraf
- Centre d’Immunologie et des Maladies Infectieuses, Inserm, CNRS, Sorbonne Université, U1135, ERL8255, CIMI-Paris, F-75013 Paris, France
| | - Nadia Amanzougaghene
- Centre d’Immunologie et des Maladies Infectieuses, Inserm, CNRS, Sorbonne Université, U1135, ERL8255, CIMI-Paris, F-75013 Paris, France
| | - Shahin Tajeri
- Centre d’Immunologie et des Maladies Infectieuses, Inserm, CNRS, Sorbonne Université, U1135, ERL8255, CIMI-Paris, F-75013 Paris, France
| | - Ayola A. Adegnika
- Centre de Recherches Médicales de Lambaréné, Lambaréné 1437, Gabon
- Institute for Tropical Medicine, University of Tübingen, 72074 Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, 38124 Braunschweig, Germany
| | - Dominique Mazier
- Centre d’Immunologie et des Maladies Infectieuses, Inserm, CNRS, Sorbonne Université, U1135, ERL8255, CIMI-Paris, F-75013 Paris, France
| | - Steffen Borrmann
- Centre de Recherches Médicales de Lambaréné, Lambaréné 1437, Gabon
- Institute for Tropical Medicine, University of Tübingen, 72074 Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, 38124 Braunschweig, Germany
- Correspondence: ; Tel.: +49-707129-85428
| |
Collapse
|
35
|
Saucedo O, Tien JH. Host movement, transmission hot spots, and vector-borne disease dynamics on spatial networks. Infect Dis Model 2022; 7:742-760. [PMID: 36439402 PMCID: PMC9672958 DOI: 10.1016/j.idm.2022.10.006] [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: 04/12/2022] [Revised: 09/04/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
We examine how spatial heterogeneity combines with mobility network structure to influence vector-borne disease dynamics. Specifically, we consider a Ross-Macdonald-type disease model on n spatial locations that are coupled by host movement on a strongly connected, weighted, directed graph. We derive a closed form approximation to the domain reproduction number using a Laurent series expansion, and use this approximation to compute sensitivities of the basic reproduction number to model parameters. To illustrate how these results can be used to help inform mitigation strategies, as a case study we apply these results to malaria dynamics in Namibia, using published cell phone data and estimates for local disease transmission. Our analytical results are particularly useful for understanding drivers of transmission when mobility sinks and transmission hot spots do not coincide.
Collapse
Affiliation(s)
- Omar Saucedo
- Department of Mathematics, Virginia Tech, Blacksburg, VA, USA
| | - Joseph H. Tien
- Department of Mathematics, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
36
|
Kamiya T, Paton DG, Catteruccia F, Reece SE. Targeting malaria parasites inside mosquitoes: ecoevolutionary consequences. Trends Parasitol 2022; 38:1031-1040. [PMID: 36209032 PMCID: PMC9815470 DOI: 10.1016/j.pt.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 11/05/2022]
Abstract
Proof-of-concept studies demonstrate that antimalarial drugs designed for human treatment can also be applied to mosquitoes to interrupt malaria transmission. Deploying a new control tool is ideally undertaken within a stewardship programme that maximises a drug's lifespan by minimising the risk of resistance evolution and slowing its spread once emerged. We ask: what are the epidemiological and evolutionary consequences of targeting parasites within mosquitoes? Our synthesis argues that targeting parasites inside mosquitoes (i) can be modelled by readily expanding existing epidemiological frameworks; (ii) provides a functionally novel control method that has potential to be more robust to resistance evolution than targeting parasites in humans; and (iii) could extend the lifespan and clinical benefit of antimalarials used exclusively to treat humans.
Collapse
Affiliation(s)
- Tsukushi Kamiya
- Centre for Interdisciplinary Research in Biology, Collège de France, Paris, France; HRB Clinical Research Facility, National University of Ireland, Galway, Ireland; Institute of Ecology and Evolution, and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK.
| | - Douglas G Paton
- Department of Immunology and Infectious Disease, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Disease, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA; Howard Hughes Medical Institute, Boston, MA, USA
| | - Sarah E Reece
- Institute of Ecology and Evolution, and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
37
|
Russell TJ, De Silva EK, Crowley VM, Shaw-Saliba K, Dube N, Josling G, Pasaje CFA, Kouskoumvekaki I, Panagiotou G, Niles JC, Jacobs-Lorena M, Denise Okafor C, Gamo FJ, Llinás M. Inhibitors of ApiAP2 protein DNA binding exhibit multistage activity against Plasmodium parasites. PLoS Pathog 2022; 18:e1010887. [PMID: 36223427 PMCID: PMC9591056 DOI: 10.1371/journal.ppat.1010887] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/24/2022] [Accepted: 09/17/2022] [Indexed: 11/06/2022] Open
Abstract
Plasmodium parasites are reliant on the Apicomplexan AP2 (ApiAP2) transcription factor family to regulate gene expression programs. AP2 DNA binding domains have no homologs in the human or mosquito host genomes, making them potential antimalarial drug targets. Using an in-silico screen to dock thousands of small molecules into the crystal structure of the AP2-EXP (Pf3D7_1466400) AP2 domain (PDB:3IGM), we identified putative AP2-EXP interacting compounds. Four compounds were found to block DNA binding by AP2-EXP and at least one additional ApiAP2 protein. Our top ApiAP2 competitor compound perturbs the transcriptome of P. falciparum trophozoites and results in a decrease in abundance of log2 fold change > 2 for 50% (46/93) of AP2-EXP target genes. Additionally, two ApiAP2 competitor compounds have multi-stage anti-Plasmodium activity against blood and mosquito stage parasites. In summary, we describe a novel set of antimalarial compounds that interact with AP2 DNA binding domains. These compounds may be used for future chemical genetic interrogation of ApiAP2 proteins or serve as starting points for a new class of antimalarial therapeutics.
Collapse
Affiliation(s)
- Timothy James Russell
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Eukaryotic Gene Regulation (CEGR), Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Malaria Research (CMaR), Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Infectious Disease Dynamics, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Erandi K. De Silva
- Lewis-Singler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
| | - Valerie M. Crowley
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Kathryn Shaw-Saliba
- Department of Molecular Biology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Namita Dube
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Gabrielle Josling
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Malaria Research (CMaR), Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Infectious Disease Dynamics, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Charisse Flerida A. Pasaje
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Irene Kouskoumvekaki
- Department of Systems Biology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Gianni Panagiotou
- Systems Biology and Bioinformatics, Leibniz Institute for Natural Products Research and Infection Biology, Hans Knöll Institute, Jena, Germany
- Department of Medicine, the University of Hong Kong, Hong Kong SAR, China
| | - Jacquin C. Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Marcelo Jacobs-Lorena
- Department of Molecular Biology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - C. Denise Okafor
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
- Department of Chemistry, Pennsylvania State University, State College, Pennsylvania, United States of America
| | | | - Manuel Llinás
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Eukaryotic Gene Regulation (CEGR), Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Malaria Research (CMaR), Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Infectious Disease Dynamics, Pennsylvania State University, State College, Pennsylvania, United States of America
- Department of Chemistry, Pennsylvania State University, State College, Pennsylvania, United States of America
| |
Collapse
|
38
|
Barillas-Mury C, Ribeiro JMC, Valenzuela JG. Understanding pathogen survival and transmission by arthropod vectors to prevent human disease. Science 2022; 377:eabc2757. [PMID: 36173836 DOI: 10.1126/science.abc2757] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Many endemic poverty-associated diseases, such as malaria and leishmaniasis, are transmitted by arthropod vectors. Pathogens must interact with specific molecules in the vector gut, the microbiota, and the vector immune system to survive and be transmitted. The vertebrate host, in turn, is infected when the pathogen and vector-derived factors, such as salivary proteins, are delivered into the skin by a vector bite. Here, we review recent progress in our understanding of the biology of pathogen transmission from the human to the vector and back, from the vector to the host. We also highlight recent advances in the biology of vector-borne disease transmission, which have translated into additional strategies to prevent human disease by either reducing vector populations or by disrupting their ability to transmit pathogens.
Collapse
Affiliation(s)
- Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - José M C Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| |
Collapse
|
39
|
Kanai M, Hagenah LM, Ashley EA, Chibale K, Fidock DA. Keystone Malaria Symposium 2022: a vibrant discussion of progress made and challenges ahead from drug discovery to treatment. Trends Parasitol 2022; 38:711-718. [PMID: 35864072 PMCID: PMC9631389 DOI: 10.1016/j.pt.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 10/17/2022]
Abstract
In recent years, the field of malaria research has made substantial progress in the areas of antimalarial drug resistance and discovery. These efforts are essential to combatting the devastating impact of malaria, which, in 2020, resulted in an estimated 241 million cases and 627 000 deaths. Recent advances in this area were presented at a Keystone Symposium entitled ‘Malaria: Confronting Challenges from Drug Discovery to Treatment’, held in person in Breckenridge, Colorado, in April 2022. Herein, we present a summary of the proceedings of this vibrant scientific exchange, which brought together a superb group of faculty, postdocs, and students from around the globe.
Collapse
Affiliation(s)
- Mariko Kanai
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Laura M Hagenah
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Elizabeth A Ashley
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Laos.
| | - Kelly Chibale
- Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa; South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch, South Africa; Department of Chemistry, University of Cape Town, Rondebosch, South Africa.
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
40
|
Muema JM, Bargul JL, Obonyo MA, Njeru SN, Matoke-Muhia D, Mutunga JM. Contemporary exploitation of natural products for arthropod-borne pathogen transmission-blocking interventions. Parasit Vectors 2022; 15:298. [PMID: 36002857 PMCID: PMC9404607 DOI: 10.1186/s13071-022-05367-8] [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: 02/24/2022] [Accepted: 06/16/2022] [Indexed: 11/26/2022] Open
Abstract
An integrated approach to innovatively counter the transmission of various arthropod-borne diseases to humans would benefit from strategies that sustainably limit onward passage of infective life cycle stages of pathogens and parasites to the insect vectors and vice versa. Aiming to accelerate the impetus towards a disease-free world amid the challenges posed by climate change, discovery, mindful exploitation and integration of active natural products in design of pathogen transmission-blocking interventions is of high priority. Herein, we provide a review of natural compounds endowed with blockade potential against transmissible forms of human pathogens reported in the last 2 decades from 2000 to 2021. Finally, we propose various translational strategies that can exploit these pathogen transmission-blocking natural products into design of novel and sustainable disease control interventions. In summary, tapping these compounds will potentially aid in integrated combat mission to reduce disease transmission trends.
Collapse
Affiliation(s)
- Jackson M Muema
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), P.O. Box 62000, Nairobi, 00200, Kenya.
| | - Joel L Bargul
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), P.O. Box 62000, Nairobi, 00200, Kenya.,International Centre of Insect Physiology and Ecology (Icipe), P.O. Box 30772, Nairobi, 00100, Kenya
| | - Meshack A Obonyo
- Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536, Egerton, 20115, Kenya
| | - Sospeter N Njeru
- Centre for Traditional Medicine and Drug Research (CTMDR), Kenya Medical Research Institute (KEMRI), P.O. Box 54840, Nairobi, 00200, Kenya
| | - Damaris Matoke-Muhia
- Centre for Biotechnology Research Development (CBRD), Kenya Medical Research Institute (KEMRI), P.O. Box 54840, Nairobi, 00200, Kenya
| | - James M Mutunga
- Department of Biological Sciences, Mount Kenya University (MKU), P.O. Box 54, Thika, 01000, Kenya.,School of Engineering Design, Technology and Professional Programs, Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
41
|
Larvicidal property of the extracts of the seaweeds; Sargassum wightii, S. ilicifolium and Gelidiella acerosa against Anopheles stephensi, Aedes aegypti and Culex quinquefasciatus. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2022. [DOI: 10.1016/j.bcab.2022.102436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
42
|
Kehrer J, Formaglio P, Muthinja JM, Weber S, Baltissen D, Lance C, Ripp J, Grech J, Meissner M, Funaya C, Amino R, Frischknecht F. Plasmodium
sporozoite disintegration during skin passage limits malaria parasite transmission. EMBO Rep 2022; 23:e54719. [PMID: 35403820 PMCID: PMC9253755 DOI: 10.15252/embr.202254719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/25/2022] Open
Abstract
During transmission of malaria‐causing parasites from mosquitoes to mammals, Plasmodium sporozoites migrate rapidly in the skin to search for a blood vessel. The high migratory speed and narrow passages taken by the parasites suggest considerable strain on the sporozoites to maintain their shape. Here, we show that the membrane‐associated protein, concavin, is important for the maintenance of the Plasmodium sporozoite shape inside salivary glands of mosquitoes and during migration in the skin. Concavin‐GFP localizes at the cytoplasmic periphery and concavin(−) sporozoites progressively round up upon entry of salivary glands. Rounded concavin(−) sporozoites fail to pass through the narrow salivary ducts and are rarely ejected by mosquitoes, while normally shaped concavin(−) sporozoites are transmitted. Strikingly, motile concavin(−) sporozoites disintegrate while migrating through the skin leading to parasite arrest or death and decreased transmission efficiency. Collectively, we suggest that concavin contributes to cell shape maintenance by riveting the plasma membrane to the subtending inner membrane complex. Interfering with cell shape maintenance pathways might hence provide a new strategy to prevent a malaria infection.
Collapse
Affiliation(s)
- Jessica Kehrer
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
- Infectious Diseases Imaging Platform Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
| | - Pauline Formaglio
- Malaria Infection and Immunity Unit Department of Parasites and Insect Vectors Institut Pasteur Paris France
| | - Julianne Mendi Muthinja
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
| | - Sebastian Weber
- Electron Microscopy Core Facility Heidelberg University Heidelberg Germany
| | - Danny Baltissen
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
| | - Christopher Lance
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
| | - Johanna Ripp
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
| | - Janessa Grech
- Experimental Parasitology Ludwig Maximilian University Munich Planegg‐Martinsried Germany
| | - Markus Meissner
- Experimental Parasitology Ludwig Maximilian University Munich Planegg‐Martinsried Germany
| | - Charlotta Funaya
- Electron Microscopy Core Facility Heidelberg University Heidelberg Germany
| | - Rogerio Amino
- Malaria Infection and Immunity Unit Department of Parasites and Insect Vectors Institut Pasteur Paris France
| | - Friedrich Frischknecht
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg Heidelberg Germany
| |
Collapse
|
43
|
Chemical and Pharmacological Properties of Decoquinate: A Review of Its Pharmaceutical Potential and Future Perspectives. Pharmaceutics 2022; 14:pharmaceutics14071383. [PMID: 35890280 PMCID: PMC9315532 DOI: 10.3390/pharmaceutics14071383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/16/2022] Open
Abstract
Decoquinate (DQ) is an antimicrobial agent commonly used as a feed additive for birds for human consumption. Its use as an additive is well established, but DQ has the potential for therapy as an antimicrobial drug for veterinary treatment and its optimized derivatives and/or formulations, mainly nanoformulations, have antimicrobial activity against pathogens that infect humans. However, DQ has a high partition coefficient and low solubility in aqueous fluids, and these biopharmaceutical properties have limited its use in humans. In this review, we highlight the antimicrobial activity and pharmacokinetic properties of DQ and highlight the solutions currently under investigation to overcome these drawbacks. A literature search was conducted focusing on the use of decoquinate against various infectious diseases in humans and animals. The search was conducted in several databases, including scientific and patent databases. Pharmaceutical nanotechnology and medicinal chemistry are the tools of choice to achieve human applications, and most of these applications have been able to improve the biopharmaceutical properties and pharmacokinetic profile of DQ. Based on the results presented here, DQ prototypes could be tested in clinical trials for human application in the coming years.
Collapse
|
44
|
van der Watt ME, Reader J, Birkholtz LM. Adapt or Die: Targeting Unique Transmission-Stage Biology for Malaria Elimination. Front Cell Infect Microbiol 2022; 12:901971. [PMID: 35755845 PMCID: PMC9218253 DOI: 10.3389/fcimb.2022.901971] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/06/2022] [Indexed: 12/25/2022] Open
Abstract
Plasmodium parasites have a complex life cycle that includes development in the human host as well as the Anopheles vector. Successful transmission of the parasite between its host and vector therefore requires the parasite to balance its investments in asexual replication and sexual reproduction, varying the frequency of sexual commitment to persist within the human host and generate future opportunities for transmission. The transmission window is extended further by the ability of stage V gametocytes to circulate in peripheral blood for weeks, whereas immature stage I to IV gametocytes sequester in the bone marrow and spleen until final maturation. Due to the low gametocyte numbers in blood circulation and with the ease of targeting such life cycle bottlenecks, transmission represents an efficient target for therapeutic intervention. The biological process of Plasmodium transmission is a multistage, multifaceted process and the past decade has seen a much deeper understanding of the molecular mechanisms and regulators involved. Clearly, specific and divergent processes are used during transmission compared to asexual proliferation, which both poses challenges but also opportunities for discovery of transmission-blocking antimalarials. This review therefore presents an update of our molecular understanding of gametocyte and gamete biology as well as the status of transmission-blocking activities of current antimalarials and lead development compounds. By defining the biological components associated with transmission, considerations for the development of new transmission-blocking drugs to target such untapped but unique biology is suggested as an important, main driver for transmission-blocking drug discovery.
Collapse
Affiliation(s)
- Mariëtte E van der Watt
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | - Janette Reader
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa.,Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Lyn-Marié Birkholtz
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa.,Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
45
|
Paton DG, Probst AS, Ma E, Adams KL, Shaw WR, Singh N, Bopp S, Volkman SK, Hien DFS, Paré PSL, Yerbanga RS, Diabaté A, Dabiré RK, Lefèvre T, Wirth DF, Catteruccia F. Using an antimalarial in mosquitoes overcomes Anopheles and Plasmodium resistance to malaria control strategies. PLoS Pathog 2022; 18:e1010609. [PMID: 35687594 PMCID: PMC9223321 DOI: 10.1371/journal.ppat.1010609] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/23/2022] [Accepted: 05/20/2022] [Indexed: 11/18/2022] Open
Abstract
The spread of insecticide resistance in Anopheles mosquitoes and drug resistance in Plasmodium parasites is contributing to a global resurgence of malaria, making the generation of control tools that can overcome these roadblocks an urgent public health priority. We recently showed that the transmission of Plasmodium falciparum parasites can be efficiently blocked when exposing Anopheles gambiae females to antimalarials deposited on a treated surface, with no negative consequences on major components of mosquito fitness. Here, we demonstrate this approach can overcome the hurdles of insecticide resistance in mosquitoes and drug resistant in parasites. We show that the transmission-blocking efficacy of mosquito-targeted antimalarials is maintained when field-derived, insecticide resistant Anopheles are exposed to the potent cytochrome b inhibitor atovaquone, demonstrating that this drug escapes insecticide resistance mechanisms that could potentially interfere with its function. Moreover, this approach prevents transmission of field-derived, artemisinin resistant P. falciparum parasites (Kelch13 C580Y mutant), proving that this strategy could be used to prevent the spread of parasite mutations that induce resistance to front-line antimalarials. Atovaquone is also highly effective at limiting parasite development when ingested by mosquitoes in sugar solutions, including in ongoing infections. These data support the use of mosquito-targeted antimalarials as a promising tool to complement and extend the efficacy of current malaria control interventions. Effective control of malaria is hampered by resistance to vector-targeted insecticides and parasite-targeted drugs. This situation is exacerbated by a critical lack of chemical diversity in both interventions and, as such, new interventions are urgently needed. Recent laboratory studies have shown that an alternative approach based on treating Anopheles mosquitoes directly with antimalarial compounds can make mosquitoes incapable of transmitting the Plasmodium parasites that cause malaria. While promising, showing that mosquito-targeted antimalarials remain effective against wild parasites and mosquitoes, including drug- and insecticide-resistant populations in malaria-endemic countries, is crucial to the future viability of this approach. In this study, carried out in the US and Burkina Faso, we show that insecticide-resistance mechanisms found in highly resistant, natural Anopheles mosquito populations do not interfere with the transmission blocking activity of tarsal exposure to the antimalarial atovaquone, and that mosquito-targeted antimalarial exposure can block transmission of parasites resistant to the main therapeutic antimalarial drug artemisinin. By combining lab, and field-based studies in this way we have demonstrated that this novel approach can be effective in areas where conventional control measures are no longer as effective.
Collapse
Affiliation(s)
- Douglas G. Paton
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
- * E-mail: (DGP); (FC)
| | - Alexandra S. Probst
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Erica Ma
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Kelsey L. Adams
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - W. Robert Shaw
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Naresh Singh
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Selina Bopp
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Sarah K. Volkman
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Domombele F. S. Hien
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Prislaure S. L. Paré
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Rakiswendé S. Yerbanga
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Abdoullaye Diabaté
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Roch K. Dabiré
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Thierry Lefèvre
- MIVEGEC, IRD, CNRS, University of Montpellier, Montpellier, France
- Laboratoire mixte international sur les vecteurs (LAMIVECT), Bobo Dioulasso, Burkina Faso
- Centre de Recherche en Écologie et Évolution de la Santé (CREES), Montpellier, France
| | - Dyann F. Wirth
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, United States of America
- * E-mail: (DGP); (FC)
| |
Collapse
|
46
|
Abstract
Some antimalarial drugs that have lost clinical usefulness have been repurposed for experimental applications. One example is sulfadiazine, an analog of p-aminobenzoic acid (pABA), which inhibits the parasite's folate synthesis pathway to block DNA synthesis. Sulfadiazine treatment of mice infected with Plasmodium yoelii and P. berghei is routinely used to enrich for gametocytes by killing asexual blood-stage parasites, but it is not well known if there are downstream effects on transmission. To determine if there was a significant effect of sulfadiazine exposure upon transmission, we transmitted Plasmodium yoelii (17XNL strain) parasites to Anopheles stephensi mosquitoes and evaluated the prevalence and intensity of infection under different sulfadiazine treatment conditions. We observed that there was a reduction in both the number of mosquitoes that became infected and in the intensity of infection if parasites were exposed to sulfadiazine in the mouse host or mosquito vector. Sulfadiazine treatment could be marginally overcome if mosquitoes were provided fresh pABA. In contrast, we determined that gametocytes exposed to sulfadiazine could develop into morphologically mature ookinetes in vitro, thus sulfadiazine exposure in the host may be reversible if the drug is washed out and the parasites are supplemented with pABA in the culture media. Overall, this indicates that sulfadiazine dampens host-to-vector transmission and that this inhibition can only be partially overcome by exposure to fresh pABA in vivo and in vitro. Because gametocytes are of great interest for developing transmission-blocking interventions, we recommend the use of less disruptive approaches for gametocyte enrichment. IMPORTANCE In this work, we have uncovered a substantial problem with how many studies of the sexual stages of rodent malaria parasites are conducted. Briefly, the isolation of sexual blood-stage Plasmodium parasites, or gametocytes, is essential to study pretransmission and transmission-stage biology of malaria. A routine method for the isolation of this specific stage in rodent-infectious malaria models is drug treatment with sulfadiazine, an antifolate that selectively kills actively replicating asexual blood-stage parasites but not gametocytes. Thus, researchers use this as a convenient way to produce highly enriched gametocyte samples. However, in this work, we describe how this standard drug selection with sulfadiazine not only kills asexual blood-stage parasites but also substantially impacts host-to-vector transmission.
Collapse
|
47
|
Erriah B, Zhu X, Hu CT, Kahr BE, Shtukenberg A, Ward MD. Crystallography of Contemporary Contact Insecticides. INSECTS 2022; 13:insects13030292. [PMID: 35323590 PMCID: PMC8949367 DOI: 10.3390/insects13030292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 03/12/2022] [Indexed: 12/04/2022]
Abstract
The active forms of contact insecticides used for combatting mosquito-borne infectious diseases are typically crystalline solids. Numerous molecular crystals are polymorphic, crystallizing in several solid forms characterized by different physicochemical properties, including bioavailability. Our laboratory recently found that the activity of crystalline contact insecticides is inversely dependent on the thermodynamic stability of their polymorphs, suggesting that efficacy can be enhanced by the manipulation of the solid-state structure. This paper argues that crystallography should be central to the development of contact insecticides, particularly because their efficacy continues to be compromised by insecticide resistance, especially among Anopheles mosquito populations that spread malaria. Although insecticidal compounds with new modes of action have been introduced to overcome resistance, new insecticides are expensive to develop and implement. The repurposing of existing chemical agents in metastable, more active crystalline forms provides an inexpensive and efficient method for ‘evergreening’ compounds whose risks are already well-established. We report herein seven new single-crystal structures of insecticides used for controlling infectious disease vectors. The structures reported herein include pyrethroid insecticides recommended by the WHO for indoor residual spraying (IRS)-bifenthrin, β-cyfluthrin, etofenprox, α-cypermethrin, and λ-cyhalothrin as well as the neonicotinoid insecticide thiacloprid.
Collapse
Affiliation(s)
| | | | | | - Bart E. Kahr
- Correspondence: (B.E.K.); (M.D.W.); Tel.: +1-212-992-9579 (B.E.K.)
| | | | - Michael D. Ward
- Correspondence: (B.E.K.); (M.D.W.); Tel.: +1-212-992-9579 (B.E.K.)
| |
Collapse
|
48
|
Consalvi S, Tammaro C, Appetecchia F, Biava M, Poce G. Malaria transmission blocking compounds: a patent review. Expert Opin Ther Pat 2022; 32:649-666. [PMID: 35240899 DOI: 10.1080/13543776.2022.2049239] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite substantial progress in the field, malaria remains a global health issue and currently available control strategies are not sufficient to achieve eradication. Agents able to prevent transmission are likely to have a strong impact on malaria control and have been prioritized as a primary objective to reduce the number of secondary infections. Therefore, there is an increased interest in finding novel drugs targeting sexual stages of Plasmodium and innovative methods to target malaria transmission from host to vector, and vice versa. AREAS COVERED This review covers innovative transmission-blocking inventions patented between 2015 and October 2021. The focus is on chemical interventions which could be used as "chemical vaccines" to prevent transmission (small molecules, carbohydrates, and polypeptides). EXPERT OPINION Even though the development of novel strategies to block transmission still requires fundamental additional research and a deeper understanding of parasite sexual stages biology, the research in this field has significantly accelerated. Among innovative inventions patented over the last six years, the surface-delivery of antimalarial drugs to kill transmission-stages parasites in mosquitoes holds the highest promise for success in malaria control strategies, opening completely new scenarios in malaria transmission-blocking drug discovery.
Collapse
Affiliation(s)
- Sara Consalvi
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy
| | - Chiara Tammaro
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy
| | - Federico Appetecchia
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy
| | - Mariangela Biava
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy
| | - Giovanna Poce
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, piazzale A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
49
|
Shaw WR, Marcenac P, Catteruccia F. Plasmodium development in Anopheles: a tale of shared resources. Trends Parasitol 2022; 38:124-135. [PMID: 34548252 PMCID: PMC8758519 DOI: 10.1016/j.pt.2021.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Interactions between the Anopheles mosquito vector and Plasmodium parasites shape how malaria is transmitted in endemic regions. The long association of these two organisms has led to evolutionary processes that minimize fitness costs of infection and benefit both players through shared nutrient resources, parasite immune suppression, and mosquito tolerance to infection. In this review we explore recent data describing how Plasmodium falciparum, the deadliest malaria parasite, associates with one of its most important natural mosquito hosts, Anopheles gambiae, and we discuss the implications of these findings for parasite transmission and vector control strategies currently in development.
Collapse
Affiliation(s)
- W Robert Shaw
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Perrine Marcenac
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
50
|
Birkholtz LM, Alano P, Leroy D. Transmission-blocking drugs for malaria elimination. Trends Parasitol 2022; 38:390-403. [DOI: 10.1016/j.pt.2022.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 12/24/2022]
|