1
|
Agbomhere Hamed M, Ahmed Surakat O, Olukayode Ekundina V, Bolajoko Jimoh K, Ezekiel Adeogun A, Omolola Akanji N, Joshua Babalola O, Chukwunonso Eya P. Neglected Tropical Diseases and Female Infertility: Possible Pathophysiological Mechanisms. J Trop Med 2025; 2025:2126664. [PMID: 40337250 PMCID: PMC12058319 DOI: 10.1155/jotm/2126664] [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: 11/04/2024] [Accepted: 03/20/2025] [Indexed: 05/09/2025] Open
Abstract
Battling female infertility has posed a global challenge, where neglected tropical diseases (NTDs) are nonetheless a notable contributing factor. NTDs affect a variety of diseases, often of a chronic nature, which are often cited as some of the most lethal diseases operating against the most economically disadvantaged populations across the globe. The various causative agents for NTDs have been documented and could originate from a myriad of sources-from bacteria, fungi and viruses to ecto- and endoparasitic species-including but not limited to helminths and protozoa. This paper will seek to describe how NTDs influence female reproductive health, together with likely mediators. While these diseases have curable forms, their effects have gone well beyond female infertility, to major pain, disability and even mortality, particularly in poorer countries, thus causing economic hardship, reduced productivity and a pool of social stigma. NTDs adversely affect female reproductive functions through multiple mechanisms, including ROS-sensitive signalling, depression of steroidogenic markers and promotion of apoptosis. The effects also may reflect their influence on ovarian histomorphology, consequently resulting in female infertility. Current-directed studies, however, suggest a potential benefit in combining drugs for the most common NTDs as a deterrent to possible female infertility endowed by NTD infection. Nonetheless, further clinical investigations will be instrumental in elucidating the probable preventive value of combination drugs as adjuvant therapy to NTDs infections. This will provide comprehensive insight into the pathophysiological and molecular basis for the impairment of female fertility brought about by NTDs, leading to the development of preventive models to curb the adverse effects of NTDs on female reproductive health. Therefore, attention should be given to providing the right, timely and effective mode of treatment for NTDs-related female infertility.
Collapse
Affiliation(s)
- Moses Agbomhere Hamed
- Department of Neuroendocrinology, The Brainwill Laboratory, Osogbo, Osun State, Nigeria
- Department of Medical Laboratory Science, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Olabanji Ahmed Surakat
- Department of Zoology, Faculty of Basic and Applied Sciences, Osun State University, Osogbo, Osun State, Nigeria
| | | | - Kabirat Bolajoko Jimoh
- Department of Neuroendocrinology, The Brainwill Laboratory, Osogbo, Osun State, Nigeria
- Department of Physiology, Faculty of Basic Medical Sciences, Osun State University, Osogbo, Osun State, Nigeria
| | - Adetomiwa Ezekiel Adeogun
- Department of Neuroendocrinology, The Brainwill Laboratory, Osogbo, Osun State, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Nafisat Omolola Akanji
- Department of Neuroendocrinology, The Brainwill Laboratory, Osogbo, Osun State, Nigeria
- Department of Physiology, University of Ibadan, Ibadan, Oyo State, Nigeria
| | | | - Patrick Chukwunonso Eya
- Department of Medical Laboratory Science, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
- Department of Environmental Health Science, National Open University of Nigeria, Jabi, Abuja, Nigeria
| |
Collapse
|
2
|
Ombura FLO, Malele I, Abd-Alla AM, Akutse KS, Ajene IJ, Khamis FM. Potential of entomopathogenic fungi for Glossina austeni control: insights into microbiome alterations and implications on sustainable management of the pest. INSECT SCIENCE 2025. [PMID: 40287897 DOI: 10.1111/1744-7917.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 04/29/2025]
Abstract
Glossina austeni Newstead (Diptera: Glossinidae) is a competent vector of the trypanosomes causing human African trypanosomiasis and the African animal trypanosomosis. Management of this pest has primarily involved trapping methods, Sterile Insect Technique, and research into vector competence-symbiotic interactions. Nevertheless, the use of entomopathogenic fungi (EPF) in integrated pest management programs for G. austeni control remains limited. Moreover, different tsetse fly species exhibit varying susceptibility to different EPF strains, indicating that no single strain is universally effective. Therefore, our study aimed to identify candidate EPF isolates for G. austeni management, evaluate the effects of temperature on the radial growth of these potent isolates, and assess the impact of the candidate EPF on the gut microbiome of G. austeni. Consequently, 16 Metarhizium anisopliae (Metschn.) Sorokin isolates were screened against G. austeni using dry conidia in an infection chamber, with the most virulent isolates having LT50 values of 3.95-9.37 d. Temperature significantly influenced the radial growth, conidia germination, and yield of these strains. There were also significant differences in conidia acquisition, retention and transmission between male and female G. austeni flies. Furthermore, all conidia receivers carried sufficient conidia, 5 d post-interaction with EPF-challenged conidia donors. Microbiome analysis revealed Wigglesworthia, Serratia, Klebsiella, and Escherichia as the most abundant taxa. Among the M. anisopliae isolates, ICIPE 82 exhibited the fastest radial growth and highest thermostability, hence selected as a potential biopesticide candidate for managing G. austeni. This study demonstrates the efficacy and potential of M. anisopliae ICIPE 82 as a biopesticide for controlling G. austeni.
Collapse
Affiliation(s)
| | - Imna Malele
- Tanzania Veterinary Laboratory Agency, Dar es Salaam, Tanzania
| | - Adly Mm Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Komivi Senyo Akutse
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Unit of Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Inusa Jacob Ajene
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Fathiya Mbarak Khamis
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Department of Zoology and Entomology, University of Pretoria, Hatfield, South Africa
| |
Collapse
|
3
|
Souza Tada da Cunha P, Rodriguez Gini AL, Man Chin C, dos Santos JL, Benito Scarim C. Recent Progress in Thiazole, Thiosemicarbazone, and Semicarbazone Derivatives as Antiparasitic Agents Against Trypanosomatids and Plasmodium spp. Molecules 2025; 30:1788. [PMID: 40333793 PMCID: PMC12029465 DOI: 10.3390/molecules30081788] [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: 03/24/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025] Open
Abstract
Neglected tropical diseases (NTDs), including Chagas disease, human African trypanosomiasis (HAT), leishmaniasis, and malaria, remain a major global health challenge, disproportionately affecting low-income populations. Current therapies for these diseases suffer from significant limitations, such as reduced efficacy, high toxicity, and emerging parasite resistance, highlighting the urgent need for new therapeutic strategies. In response, substantial efforts have been directed toward the synthesis of new molecules with improved potency, selectivity, and pharmacokinetic profiles. However, despite many of these compounds exhibiting favorable ADMET (absorption, distribution, metabolism, excretion, and toxicity) profiles and strong in vitro activity, their translation into in vivo models remains limited. Key challenges include the lack of investment, the absence of fully representative experimental models, and difficulties in extrapolating cell-based assay results to more complex biological systems. In this review, we analyzed the latest advancements (2019-2024) in the development of these compound classes, correlating predictive parameters with their observed biological activity. Among these parameters, we highlighted the partition coefficient (LogP), which measures a compound's lipophilicity and influences its ability to cross biological membranes, and Caco-2 cell permeability, an in vitro model widely used to predict intestinal drug absorption. Additionally, we prioritized the most promising molecules and structural classes for pharmaceutical development, discussing structure-activity relationships (SARs) and the remaining challenges that must be overcome to enable the clinical application of these compounds in the treatment of NTDs.
Collapse
Affiliation(s)
| | | | | | | | - Cauê Benito Scarim
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (P.S.T.d.C.); (A.L.R.G.); (C.M.C.); (J.L.d.S.)
| |
Collapse
|
4
|
Konan YJR, Ta BTD, Berté D, Coulibaly B, Coulibaly KD, Egnankon NS, Diarrassouba F, Djabo KA, Watier-Grillot S, Demoncheaux JP, De Marie Kouadio KA, N'Dri L, Solano P, Ravel S, Adingra GP, Barreaux A, Ségard A, Kaba D, Jamonneau V, De Meeûs T, Djohan V. Challenges and lessons from a vector control campaign targeting Glossina palpalis palpalis in an isolated protected forest area in Abidjan, Côte d'Ivoire. Parasite 2025; 32:25. [PMID: 40239040 PMCID: PMC12002674 DOI: 10.1051/parasite/2025017] [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: 06/03/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Vector control (VC) is one of the strategies employed to manage African trypanosomoses. This study aimed at assessing the effectiveness of a VC campaign against Glossina palpalis palpalis using tiny targets (TTs) impregnated with insecticide in an isolated, protected forest in Abidjan, Côte d'Ivoire, while considering ecological, genetic, and operational factors. Between January 2020 and September 2022, 2,712 TTs were deployed at 684 sites, covering a total area of 1.7 km2. VC monitoring was conducted using Vavoua traps during 12 evaluation surveys, between June 2020 and March 2023. Five months after the initial TT deployment, tsetse fly density had decreased by 98.53%. Although tsetse density remained low due to TT redeployment and reinforcement, there was a significant increase a few months after the last redeployment. VC appeared to have minimal impact on the genetic structuring of G. p. palpalis. This suggested recruitment of local surviving tsetse flies all along the VC campaign due to a low probability of tsetse coming into contact with TTs, or to the evolution of behavioral or physiological resistance to control efforts. The genetic study revealed that one of the microsatellite markers used, the GPCAG locus, exhibited a selection signature possibly in response to VC. This could partly explain the challenges encountered in eliminating a seemingly isolated tsetse population thriving in a particularly favorable habitat.
Collapse
Affiliation(s)
- Yao Jean Rodrigue Konan
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire - Université Felix Houphouët-Boigny, 01 BPV 34, Abidjan, Côte d'Ivoire
| | - Bi Tra Dieudonné Ta
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire
| | - Djakaridja Berté
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire
| | - Bamoro Coulibaly
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire
| | - Kinifo Donatien Coulibaly
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire - Université Peleforo Gon Coulibaly, BP 1328 Korhogo, Côte d'Ivoire
| | - Nick Steven Egnankon
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire - Université Jean Lorougnon Guédé, BP 150, Daloa, Côte d'Ivoire
| | | | - Kouassi Albert Djabo
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire
| | - Stéphanie Watier-Grillot
- Direction interarmées du service de santé pour l'Afrique Centrale et de l'Ouest, BP 175, Abidjan, Côte d'Ivoire
| | - Jean-Paul Demoncheaux
- Direction interarmées du service de santé pour l'Afrique Centrale et de l'Ouest, BP 175, Abidjan, Côte d'Ivoire
| | | | - Louis N'Dri
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire
| | - Philippe Solano
- Intertryp, Université de Montpellier, Cirad, IRD, TA A-17/G, Campus International de Baillarguet, 34398, Montpellier Cedex 5, France
| | - Sophie Ravel
- Intertryp, Université de Montpellier, Cirad, IRD, TA A-17/G, Campus International de Baillarguet, 34398, Montpellier Cedex 5, France
| | - Guy Pacôme Adingra
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire
| | - Antoine Barreaux
- Intertryp, Université de Montpellier, Cirad, IRD, TA A-17/G, Campus International de Baillarguet, 34398, Montpellier Cedex 5, France - Animal health Theme, International Centre of Insect Physiology and Ecology (ICIPE), PO Box 30772-00100, Nairobi, Kenya
| | - Adeline Ségard
- Intertryp, Université de Montpellier, Cirad, IRD, TA A-17/G, Campus International de Baillarguet, 34398, Montpellier Cedex 5, France
| | - Dramane Kaba
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire
| | - Vincent Jamonneau
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire - Intertryp, Université de Montpellier, Cirad, IRD, TA A-17/G, Campus International de Baillarguet, 34398, Montpellier Cedex 5, France
| | - Thierry De Meeûs
- Intertryp, Université de Montpellier, Cirad, IRD, TA A-17/G, Campus International de Baillarguet, 34398, Montpellier Cedex 5, France
| | - Vincent Djohan
- Institut Pierre Richet, Institut National de Sante Publique, 01 BP 1500, Bouaké, Côte d'Ivoire - Université Felix Houphouët-Boigny, 01 BPV 34, Abidjan, Côte d'Ivoire
| |
Collapse
|
5
|
Fiorenza G, Piccinno R, Bruzzese DJ, Scolari F, Milanesi G, Casali C, Gomulski LM, Lescai F, Forneris F, Gasperi G, Dera KSM, de Beer C, Abd-Alla AMM, Aksoy S, Malacrida AR. Effect of Spiroplasma infection on the mating behavior of Glossina fuscipes fuscipes. INSECT SCIENCE 2025. [PMID: 40195916 DOI: 10.1111/1744-7917.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 04/09/2025]
Abstract
Tsetse flies are insects of significant public health and zoonotic importance as they are the main vectors of African trypanosomes. To date, an effective vaccine is unavailable and efforts to limit the spread of the disease primarily rely on controlling the tsetse populations. The discovery of Spiroplasma (class Mollicutes) in Glossina fuscipes fuscipes (Gff) (palpalis subgroup), offers promising insights into its potential use as a biological control agent to hinder trypanosomes infection in tsetse flies. Indeed, a negative correlation between Spiroplasma and trypanosome co-infection has been observed. Using a laboratory strain of Gff, we provide fundamental biological insights into the effects of Spiroplasma infection on the mating behavior of the fly. We found a sex-biased Spiroplasma infection, with males exhibiting a higher infection rate. Mass mating experiments revealed a higher mating propensity in Spiroplasma-infected flies. Additionally, the presence of Spiroplasma influenced premating isolation, leading to nonrandom mating patterns that favored the pairing of individuals with matching infection statuses. Moreover, we present evidence of Spiroplasma vertical paternal transmission. By analyzing female reproductive tissues at 2 and 24 h postmating, we confirmed that an infected male can transfer Spiroplasma to the female via the spermatophore, which can subsequently migrate to the spermathecae. This study provides foundational insights into the role of Spiroplasma in tsetse fly mating behavior and provides supporting evidence for vertical transmission from infected males.
Collapse
Affiliation(s)
- Giulia Fiorenza
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Riccardo Piccinno
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Daniel J Bruzzese
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Francesca Scolari
- Institute of Molecular Genetics IGM CNR "Luigi Luca Cavalli-Sforza", Pavia, Italy
| | - Gloria Milanesi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Claudio Casali
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Ludvik M Gomulski
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Francesco Lescai
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Federico Forneris
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Giuliano Gasperi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
- Institute of Molecular Genetics IGM CNR "Luigi Luca Cavalli-Sforza", Pavia, Italy
| | - Kiswend-Sida M Dera
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, Vienna, Austria
| | - Chantel de Beer
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, Vienna, Austria
| | - Adly M M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, Vienna, Austria
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Anna R Malacrida
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
6
|
Ilboudo H, N'Djetchi MK, Kaboré WJ, Kaboré J, Traoré BM, Tahita MC, Ahouty BA, Deborggraeve S, Eloiflin R, Ségard A, Bucheton B, Koffi M, Jamonneau V. Evaluation of the AnTat A/B and LiTat A/B primers for the detection of Trypanosoma bruceigambiense. Exp Parasitol 2025; 271:108929. [PMID: 40044069 DOI: 10.1016/j.exppara.2025.108929] [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: 09/13/2024] [Revised: 01/14/2025] [Accepted: 03/02/2025] [Indexed: 03/25/2025]
Abstract
Elimination of gambiense human African trypanosomiasis (gHAT) as a public health problem has been reached or is in sight in a number of endemic foci and the next step is now to reach the elimination of transmission. The ability to detect Trypanosomabruceigambiense (T.b.gambiense) in both the last human cases and in a suspected animal reservoir becomes increasingly important to reach this goal. We have evaluated here the diagnostic performance of the AnTat A/B and LiTat A/B primers in comparison with the TBR, TgsGP and nested TgsGP PCRs that are currently used for the molecular diagnosis of gHAT. The evaluation was based on serial DNA dilutions from two T.b.gambiense strains for sensitivity, purified reference strains for specificity and field strains isolated from pigs in Côte d'Ivoire for field application. Results showed that the two PCRs (AnTat A/B and LiTat A/B) are not specific for T.b.gambiense, limiting their relevance for studies on suspected animal reservoirs. However, they could represent complementary tools to improve the molecular diagnosis of gHAT in the elimination process even if the detection limit was lowest than for the TgsGP PCR. The results also once more suggest that nested TgsGP PCR should be interpreted with caution as they may lead to an over-estimation of the T.b.gambiense prevalence particularly in animal studies.
Collapse
Affiliation(s)
- Hamidou Ilboudo
- Institut de Recherche en Sciences de la Santé/Unité de Recherche Clinique de Nanoro, CMS 11, 11 BP 218, Ouagadougou, Burkina Faso.
| | - Martial Kassi N'Djetchi
- Université Jean Lorougnon Guédé, Unité de Recherche en Génétique et Épidémiologie Moléculaire, UFR Environnement, BP 150, Daloa, Côte d'Ivoire
| | - Windingoudi Justin Kaboré
- Centre International de Recherche-Développement sur l'Elevage en zone Subhumide, 01, 01 BP 454, Bobo- Dioulasso, Burkina Faso
| | - Jacques Kaboré
- Centre International de Recherche-Développement sur l'Elevage en zone Subhumide, 01, 01 BP 454, Bobo- Dioulasso, Burkina Faso; Université Nazi Boni, Unité de Formation et de Recherche en Sciences de la Vie et de la Terre (UFR/SVT), 01 BP 1091, Bobo-Dioulasso, Burkina Faso
| | - Barkissa Mélika Traoré
- Université Jean Lorougnon Guédé, Unité de Recherche en Génétique et Épidémiologie Moléculaire, UFR Environnement, BP 150, Daloa, Côte d'Ivoire
| | - Marc Christian Tahita
- Institut de Recherche en Sciences de la Santé/Unité de Recherche Clinique de Nanoro, CMS 11, 11 BP 218, Ouagadougou, Burkina Faso
| | - Bernardin Ahouty Ahouty
- Université Jean Lorougnon Guédé, Unité de Recherche en Génétique et Épidémiologie Moléculaire, UFR Environnement, BP 150, Daloa, Côte d'Ivoire
| | - Stijn Deborggraeve
- Institute of Tropical Medicine Antwerp, Nationalestraat 155, B-2000, Antwerp, Belgium
| | - Roger Eloiflin
- Intertryp, IRD-CIRAD-University of Montpellier, TA A-17/G, Campus International de Baillarguet, F-34398, Montpellier, France
| | - Adeline Ségard
- Intertryp, IRD-CIRAD-University of Montpellier, TA A-17/G, Campus International de Baillarguet, F-34398, Montpellier, France
| | - Bruno Bucheton
- Intertryp, IRD-CIRAD-University of Montpellier, TA A-17/G, Campus International de Baillarguet, F-34398, Montpellier, France
| | - Mathurin Koffi
- Université Jean Lorougnon Guédé, Unité de Recherche en Génétique et Épidémiologie Moléculaire, UFR Environnement, BP 150, Daloa, Côte d'Ivoire
| | - Vincent Jamonneau
- Intertryp, IRD-CIRAD-University of Montpellier, TA A-17/G, Campus International de Baillarguet, F-34398, Montpellier, France
| |
Collapse
|
7
|
Pinto Torres JE, Claes M, Hendrickx R, Yuan M, Smiejkowska N, Van Wielendaele P, Hacisuleyman A, De Winter H, Muyldermans S, Michels PAM, Walkinshaw MD, Versées W, Caljon G, Magez S, Sterckx YGJ. Allosteric inhibition of trypanosomatid pyruvate kinases by a camelid single-domain antibody. eLife 2025; 13:RP100066. [PMID: 40163365 PMCID: PMC11957543 DOI: 10.7554/elife.100066] [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: 04/02/2025] Open
Abstract
African trypanosomes are the causative agents of neglected tropical diseases affecting both humans and livestock. Disease control is highly challenging due to an increasing number of drug treatment failures. African trypanosomes are extracellular, blood-borne parasites that mainly rely on glycolysis for their energy metabolism within the mammalian host. Trypanosomal glycolytic enzymes are therefore of interest for the development of trypanocidal drugs. Here, we report the serendipitous discovery of a camelid single-domain antibody (sdAb aka Nanobody) that selectively inhibits the enzymatic activity of trypanosomatid (but not host) pyruvate kinases through an allosteric mechanism. By combining enzyme kinetics, biophysics, structural biology, and transgenic parasite survival assays, we provide a proof-of-principle that the sdAb-mediated enzyme inhibition negatively impacts parasite fitness and growth.
Collapse
Affiliation(s)
- Joar Esteban Pinto Torres
- Laboratory for Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB)BrusselBelgium
| | - Mathieu Claes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| | - Rik Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| | - Meng Yuan
- School of Biological Sciences, The University of EdinburghEdinburghUnited Kingdom
| | - Natalia Smiejkowska
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| | - Pieter Van Wielendaele
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| | - Aysima Hacisuleyman
- Department of Computational Biology, University of LausanneLausanneSwitzerland
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, University of AntwerpWilrijkBelgium
| | - Serge Muyldermans
- Laboratory for Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB)BrusselBelgium
| | - Paul AM Michels
- School of Biological Sciences, The University of EdinburghEdinburghUnited Kingdom
| | - Malcolm D Walkinshaw
- School of Biological Sciences, The University of EdinburghEdinburghUnited Kingdom
| | - Wim Versées
- VIB-VUB Center for Structural Biology, VIBBrusselsBelgium
- Structural Biology Brussels, Vrije Universiteit BrusselBrusselsBelgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| | - Stefan Magez
- Laboratory for Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB)BrusselBelgium
- Center for Biomedical Research, Ghent University Global CampusIncheonRepublic of Korea
- Department for Biochemistry and Microbiology, Ghent UniversityGhentBelgium
| | - Yann G-J Sterckx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| |
Collapse
|
8
|
Barrett MP. Transforming the chemotherapy of human African trypanosomiasis. Clin Microbiol Rev 2025; 38:e0015323. [PMID: 39772631 PMCID: PMC11905362 DOI: 10.1128/cmr.00153-23] [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] [Indexed: 01/11/2025] Open
Abstract
SUMMARYPrior to 2019, when the orally available drug fexinidazole began its clinical use, the treatment of human African trypanosomiasis (HAT) was complex and unsatisfactory for many reasons. Two sub-species of the Trypanosoma brucei parasite are responsible for HAT, namely the rhodesiense form found in East and Southern Africa and the gambiense form found in Central and West Africa. Diseases caused by both forms manifest in two stages: stage 1 before and stage 2 after central nervous system involvement. Prior to 2019, different drugs were required for each of the two parasite sub-species at each stage. Gambiense disease required pentamidine or nifurtimox-eflornithine combination therapy, while for rhodesiense disease, suramin or melarsoprol was given for stages 1 and 2, respectively. These drugs all suffered complications including protracted administration regimens involving multiple injections with drug-induced adverse effects common. Today, a single drug, fexinidazole, can be given orally in most cases for both diseases at either stage. Another compound, acoziborole, effective in both stages 1 and 2 gambiense disease with a single dosing is anticipated to become available within a few years. Moreover, the recent engagement of multilateral organizations in seeking other compounds that could be used in HAT therapy has also been successful, and a rich vein of new trypanocides has been discovered. Here, the clinical use, modes of action, and resistance risks for drugs used against HAT are discussed.
Collapse
Affiliation(s)
- Michael P. Barrett
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
9
|
Liu Y, Jiang N, Zuo S, Feng Y, Chen R, Zhang Y, Zhang N, Sang X, Chen Q. Graphene quantum dots disrupt the mitochondrial potential of Trypanosoma brucei by interacting with the p18 subunit of ATP synthase F 1 after endocytosis via the VSG recycling pathway. J Colloid Interface Sci 2025; 679:975-986. [PMID: 39418900 DOI: 10.1016/j.jcis.2024.10.054] [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: 06/03/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
HYPOTHESIS Trypanosomiasis is one of the main threats to human and animal health in African countries. Trypanosoma brucei can evade the host immune recognition by rapidly altering its variant surface glycoprotein (VSG). The ATP synthase F1 subunit of the parasite exhibits extremely low similarity to that of its mammalian hosts, hypothetically making it an ideal target for the development of novel therapeutics. EXPERIMENTS Graphene quantum dots (GQDs) were synthesized, and their adhesion to T. brucei surface and internalization was observed microscopically. The activity of ATP synthase and mitochondrial membrane potential of T. brucei were measured after exposure to GQDs. Proteomics, biolayer interferometry, and molecular dynamic simulations were utilized to evaluate the interaction between GQDs with the target proteins. FINDINGS GQDs specifically adhered to the VSG of T. brucei and were conveyed inside the parasite via the VSG internalization pathway. The GQDs promoted intracellular ROS production, interacted with, and inhibited the activity of the p18 subunit of ATP synthase, disrupted parasite mitochondrial membrane potential. Additionally, the GQDs caused a decrease in aminoacyl - tRNA biosynthesis, and upregulated RNA and protein degradation pathways. The findings of this study offer a novel avenue for the target-oriented discovery of anti-trypanosome drugs.
Collapse
Affiliation(s)
- Yize Liu
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Si Zuo
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Yiwei Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Naiwen Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China.
| |
Collapse
|
10
|
Black JA, Poulton BC, Gonzaga B, Iskantar A, Paape D, Tosi LRO, McCulloch R. AUK3 is required for faithful nuclear segregation in the bloodstream form of Trypanosoma brucei. Mol Biochem Parasitol 2025; 261:111664. [PMID: 39746453 DOI: 10.1016/j.molbiopara.2024.111664] [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: 10/23/2024] [Revised: 11/28/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Eukaryotic chromosomes segregate faithfully prior to nuclear division to ensure genome stability. If segregation becomes defective, the chromosome copy number of the cell may alter leading to aneuploidy and/or polyploidy, both common hallmarks of cancers. In eukaryotes, aurora kinases regulate chromosome segregation during mitosis and meiosis, but their functions in the divergent, single-celled eukaryotic pathogen Trypanosoma brucei are less understood. Here, we focused on one of three aurora kinases in these parasites, TbAUK3, a homologue of the human aurora kinase AURKC, whose functions are primarily restricted to meiosis. We show that RNAi targeted depletion of TbAUK3 correlates with nuclear segregation defects, reduced proliferation, and decreased DNA synthesis, suggestive of a role for TbAUK3 during mitotic, not meiotic, chromosome segregation. Moreover, we uncover a putative role for TbAUK3 during the parasite's response to DNA damage since we show that depletion of TbAUK3 enhances DNA instability and sensitivity to genotoxic agents.
Collapse
Affiliation(s)
- J A Black
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; University of Glasgow, Centre for Parasitology, School of Infection and Immunity, 120 University Place, Sir Graeme Davies Building, Glasgow G12 8TA, United Kingdom
| | - B C Poulton
- University of Glasgow, Centre for Parasitology, School of Infection and Immunity, 120 University Place, Sir Graeme Davies Building, Glasgow G12 8TA, United Kingdom; Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | - B Gonzaga
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - A Iskantar
- University of Glasgow, Centre for Parasitology, School of Infection and Immunity, 120 University Place, Sir Graeme Davies Building, Glasgow G12 8TA, United Kingdom
| | - D Paape
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - L R O Tosi
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - R McCulloch
- University of Glasgow, Centre for Parasitology, School of Infection and Immunity, 120 University Place, Sir Graeme Davies Building, Glasgow G12 8TA, United Kingdom.
| |
Collapse
|
11
|
Cerone M, Smith TK. Exploring the activity of the putative Δ6-desaturase and its role in bloodstream form life-cycle transitions in Trypanosoma brucei. PLoS Pathog 2025; 21:e1012691. [PMID: 39965027 PMCID: PMC11867338 DOI: 10.1371/journal.ppat.1012691] [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: 10/22/2024] [Revised: 02/27/2025] [Accepted: 01/26/2025] [Indexed: 02/20/2025] Open
Abstract
Trypanosomatids have been shown to possess an exclusive and finely regulated biosynthetic pathway for de novo synthesis of fatty acids (FAs) and particularly of polyunsaturated fatty acids (PUFAs). The key enzymes for the process of unsaturation are known as desaturases. In this work, we explored the biocatalytic activity of the putative Δ6-desaturase (Tb11.v5.0580) in the native organism T. brucei, whose expression level varies dramatically between life cycle stages. Utilising FA analysis via GC-MS, we were able to elucidate i) via genetic manipulation of the level of expression of Δ6-desaturases in both procyclic (PCF) and bloodstream (BSF) forms of T. brucei and ii) via supplementation of the media with various levels of FA sources, that docosahexaenoic acid (22:6) and/or docosapentaenoic acid (22:5) are the products, while arachidonic acid (20:4) and/or docosatetraenoic acid (22:4) are the substrates of this Δ6-desaturase. Surprisingly, we were able to observe, via lipidomic analysis with ESI-MS/MS, an increase in inositol-phosphoryl ceramide (IPC) in response to the overexpression of Δ6-desaturase in low-fat media in BSF. The formation of IPC is normally only observed in the stumpy and procyclic forms of T. brucei. Therefore, the expression levels of Δ6-desaturases, which increases between BSF, stumpy and PCF, might be involved in the cascade(s) of metabolic events that contributes to these remodelling of the lipid pools and ultimately morphological changes, which are key to the transition between these life-cycle stages. We were in fact able to show that the overexpression of Δ6-desaturase is indeed linked to the expression of protein associated with differentiation (PAD1) in stumpy, and of the upregulation of some proteins and metabolites which are normally upregulated in stumpy and PCF.
Collapse
Affiliation(s)
- Michela Cerone
- Schools of Chemistry and Biology, Biomedical Sciences Research Complex, North Haugh, University of St Andrews, St Andrews, Scotland
| | - Terry K. Smith
- Schools of Chemistry and Biology, Biomedical Sciences Research Complex, North Haugh, University of St Andrews, St Andrews, Scotland
| |
Collapse
|
12
|
Tablado Alonso S, Biéler S, Inocêncio da Luz R, Verlé P, Büscher P, Hasker E. Retrospective clinical performance evaluation of the Abbott Bioline HAT 2.0, a rapid diagnostic test for human African trypanosomiasis based on recombinant antigens. Trop Med Int Health 2025; 30:135-142. [PMID: 39716774 PMCID: PMC11791878 DOI: 10.1111/tmi.14077] [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] [Indexed: 12/25/2024]
Abstract
BACKGROUND Rapid diagnostic tests for the serological detection of gambiense human African trypanosomiasis (gHAT) have been developed to overcome the limitations of the traditional screening method, CATT/T. b. gambiense. The Abbott Bioline human African trypanosomiasis (HAT) 2.0 rapid diagnostic test has recently been developed by Abbott using only recombinant antigens. The objective of this study was to evaluate its clinical sensitivity and specificity, in comparison with the other available rapid diagnostic tests and CATT. METHODOLOGY/PRINCIPAL FINDINGS For this study, archived plasma samples from 150 gHAT cases and 150 endemic controls originating from Chad, Guinea, the Democratic Republic of the Congo and Uganda were analysed on the following tests: CATT/T. b. gambiense, the HAT Sero K-SeT, SD BIOLINE HAT and Abbott Bioline HAT 2.0 rapid diagnostic tests, and the immune trypanolysis test. The sensitivity and specificity of Abbott Bioline HAT 2.0 were 96.7% and 78.4%, respectively, and the sensitivity and specificity of SD BIOLINE HAT were 99.3% and 74.1%. The sensitivity and specificity of CATT were 98.7% and 89.2%. The sensitivity and specificity of HAT Sero K-SeT were 99.3% and 81.3%. CONCLUSIONS/SIGNIFICANCE The sensitivity and specificity of the Abbott Bioline HAT 2.0 are comparable to those of its predecessors HAT Sero K-SeT and SD BIOLINE HAT. However, considering the decreasing prevalence of gHAT, a higher specificity of the tests used for screening is desirable to improve their positive predictive value.
Collapse
Affiliation(s)
| | - Sylvain Biéler
- Neglected Tropical Diseases ProgramFoundation for Innovative New DiagnosticsGenevaSwitzerland
| | | | - Paul Verlé
- Department of Public HealthInstitute of Tropical MedicineAntwerpBelgium
| | - Philippe Büscher
- Department of Public HealthInstitute of Tropical MedicineAntwerpBelgium
| | - Epco Hasker
- Department of Public HealthInstitute of Tropical MedicineAntwerpBelgium
| |
Collapse
|
13
|
Sharif M, Greenberg L, Bangs J. Multifunctional roles of Sec13 paralogues in the euglenozoan Trypanosoma brucei. Open Biol 2025; 15:240324. [PMID: 39999875 PMCID: PMC11858755 DOI: 10.1098/rsob.240324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 02/27/2025] Open
Abstract
Secretory cargos are exported from the ER via COPII-coated vesicles that have an inner matrix of Sec23/Sec24 heterotetramers and an outer cage of Sec13/Sec31 heterotetramers. In addition to COPII, Sec13 is part of the nuclear pore complex (NPC) and the regulatory SEA/GATOR complex in eukaryotes, which typically have one Sec13 orthologue. The kinetoplastid parasite Trypanosoma brucei has two paralogues: TbSec13.1, an accepted component of both COPII and the NPC, and TbSec13.2. Little is known about TbSec13.2, but others have proposed that it, and its orthologue in the distantly related diplonemid Paradiplonema papillatum, operate exclusively in the SEA/GATOR complex, and that this represents an evolutionary diversification of function unique to the euglenozoan protists. Using RNAi silencing in trypanosomes, we show both TbSec13s are essential. Knockdown of each dramatically and equally delays transport of GPI-anchored secretory cargo, indicating roles for both in COPII-mediated trafficking from the ER. Immunofluorescence and proximity labelling studies confirm that both TbSec13.1 and TbSec13.2 co-localize with TbSec24.1 to ER exit sites, and thus are functional components of the COPII machinery. Our findings indicate that TbSec13.2 function is not restricted to the SEA/GATOR complex in trypanosomes.
Collapse
Affiliation(s)
- Mohamed Sharif
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main Street, Buffalo, NY14203, USA
| | - Lydia Greenberg
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main Street, Buffalo, NY14203, USA
| | - James Bangs
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main Street, Buffalo, NY14203, USA
| |
Collapse
|
14
|
Sharma V, Das R, Mehta DK, Sharma D, Aman S, Khan MU. Quinolone scaffolds as potential drug candidates against infectious microbes: a review. Mol Divers 2025; 29:711-737. [PMID: 38683488 DOI: 10.1007/s11030-024-10862-4] [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: 02/16/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024]
Abstract
Prevalence of microbial infections and new rising pathogens are signified as causative agent for variety of serious and lethal health crisis in past years. Despite medical advances, bacterial and fungal infections continue to be a rising problem in the health care system. As more bacteria develop resistance to antibiotics used in therapy, and as more invasive microbial species develop resistance to conventional antimicrobial drugs. Relevant published publications from the last two decades, up to 2024, were systematically retrieved from the MEDLINE/PubMed, SCOPUS, EMBASE, and WOS databases using keywords such as quinolones, anti-infective, antibacterial, antimicrobial resistance and patents on quinolone derivatives. With an approach of considerable interest towards novel heterocyclic derivatives as novel anti-infective agents, researchers have explored these as essential tools in vistas of drug design and development. Among heterocycles, quinolones have been regarded extremely essential for the development of novel derivatives, even able to tackle the associated resistance issues. The quinolone scaffold with its bicyclic structure and specific functional groups such as the carbonyl and acidic groups, is indeed considered a valuable functionalities for further lead generation and optimization in drug discovery. Besides, the substitution at N-1, C-3 and C-7 positions also subjected to be having a significant role in anti-infective potential. In this article, we intend to highlight recent quinolone derivatives based on the SAR approach and anti-infective potential such as antibacterial, antifungal, antimalarial, antitubercular, antitrypanosomal and antiviral activities. Moreover, some recent patents granted on quinolone-containing derivatives as anti-infective agents have also been highlighted in tabular form. Due consideration of this, future research in this scaffold is expected to be useful for aspiring scientists to get pharmacologically significant leads.
Collapse
Affiliation(s)
- Vishal Sharma
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Rina Das
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Dinesh Kumar Mehta
- Department of Pharmaceutical Chemistry, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India.
| | - Diksha Sharma
- Swami Devidyal College of Pharmacy, Barwala, 134118, India
| | - Shahbaz Aman
- Department of Microbiology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - M U Khan
- Department of pharmaceutical Chemistry & Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Uniazah, Al Qassim, Saudi Arabia
| |
Collapse
|
15
|
Lindner AK, Lejon V, Barrett MP, Blumberg L, Bukachi SA, Chancey RJ, Edielu A, Matemba L, Mesha T, Mwanakasale V, Pasi C, Phiri T, Seixas J, Akl EA, Probyn K, Villanueva G, Simarro PP, Kadima Ebeja A, Franco JR, Priotto G. New WHO guidelines for treating rhodesiense human African trypanosomiasis: expanded indications for fexinidazole and pentamidine. THE LANCET. INFECTIOUS DISEASES 2025; 25:e77-e85. [PMID: 39389073 DOI: 10.1016/s1473-3099(24)00581-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024]
Abstract
Human African trypanosomiasis is a neglected tropical disease that is usually fatal without treatment. WHO has revised its rhodesiense human African trypanosomiasis treatment guidelines on the basis of an independent systematic literature review and following the GRADE methodology. This Review reports on the decision-making process and summarises the new recommendations and their potential implications for health-care professionals and policy makers. Due to data scarcity, all recommendations are conditional and based on very low certainty of evidence. Fexinidazole replaces suramin and melarsoprol as the first-line therapy in individuals aged 6 years and older with a bodyweight of 20 kg or more. As fexinidazole is effective in both stages of rhodesiense human African trypanosomiasis, a lumbar puncture for staging is no longer required. In settings in which first-choice drugs are not readily available, immediate interim treatment with pentamidine is suggested. The introduction of oral fexinidazole represents an advancement in the management of rhodesiense human African trypanosomiasis considering the life-threatening adverse reactions individuals can have to melarsoprol. However, children below the age or weight limits remain ineligible for treatment with fexinidazole.
Collapse
Affiliation(s)
- Andreas K Lindner
- Charité-Universitätsmedizin Berlin, Charité Center for Global Health, Institute of International Health, Berlin, Germany.
| | - Veerle Lejon
- Intertryp, Institut de Recherche pour le Développement, CIRAD, University of Montpellier, Montpellier, France
| | - Michael P Barrett
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Lucille Blumberg
- National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Salome A Bukachi
- Institute of Anthropology, Gender and African Studies, University of Nairobi, Nairobi, Kenya; Department of Anthropology, Durham University, Durham, UK
| | | | - Andrew Edielu
- Immunomodulation and Vaccines Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Lucas Matemba
- National Institute for Medical Research, Dodoma, Tanzania
| | | | | | | | | | - Jorge Seixas
- Institute of Hygiene and Tropical Medicine and Global Health and Tropical Medicine R&D Center, NOVA University, Lisbon, Portugal
| | - Elie A Akl
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | | | | | - Pere P Simarro
- World Health Organization, Global Neglected Tropical Diseases Programme, Prevention, Treatment and Care Unit, Geneva, Switzerland
| | - Augustin Kadima Ebeja
- World Health Organization Office for the Democratic Republic of the Congo, Kinshasa, Democratic Republic of the Congo
| | - Jose R Franco
- Neglected Tropical Diseases Department, World Health Organization, Geneva, Switzerland
| | - Gerardo Priotto
- Neglected Tropical Diseases Department, World Health Organization, Geneva, Switzerland
| |
Collapse
|
16
|
Bernhard H, Petržílková H, Popelářová B, Ziemkiewicz K, Bartosik K, Warmiński M, Tengo L, Gröger H, Dolce LG, Mackereth CD, Micura R, Jemielity J, Kowalinski E. Structural basis of Spliced Leader RNA recognition by the Trypanosoma brucei cap-binding complex. Nat Commun 2025; 16:685. [PMID: 39814716 PMCID: PMC11735809 DOI: 10.1038/s41467-024-55373-w] [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: 05/21/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025] Open
Abstract
Kinetoplastids are a clade of eukaryotic protozoans that include human parasitic pathogens like trypanosomes and Leishmania species. In these organisms, protein-coding genes are transcribed as polycistronic pre-mRNAs, which need to be processed by the coupled action of trans-splicing and polyadenylation to yield monogenic mature mRNAs. During trans-splicing, a universal RNA sequence, the spliced leader RNA (SL RNA) mini-exon, is added to the 5'-end of each mRNA. The 5'-end of this mini-exon carries a hypermethylated cap structure and is bound by a trypanosomatid-specific cap-binding complex (CBC). The function of three of the kinetoplastid CBC subunits is unknown, but an essential role in cap-binding and trans-splicing has been suggested. Here, we report cryo-EM structures that reveal the molecular architecture of the Trypanosoma brucei CBC (TbCBC) complex. We find that TbCBC interacts with two distinct features of the SL RNA. The TbCBP20 subunit interacts with the m7G cap while TbCBP66 recognizes double-stranded portions of the SL RNA. Our findings pave the way for future research on mRNA maturation in kinetoplastids. Moreover, the observed structural similarities and differences between TbCBC and the mammalian cap-binding complex will be crucial for considering the potential of TbCBC as a target for anti-trypanosomatid drug development.
Collapse
Affiliation(s)
- Harald Bernhard
- EMBL Grenoble, 71 Avenue des Martyrs, Grenoble, France
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes (UGA), Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Centre National de la Recherche Scientifique (CNRS), Grenoble, France
| | | | - Barbora Popelářová
- EMBL Grenoble, 71 Avenue des Martyrs, Grenoble, France
- Department of Experimental Biology, Section of Microbiology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | - Karolina Bartosik
- Institute of Organic Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Marcin Warmiński
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Laura Tengo
- EMBL Grenoble, 71 Avenue des Martyrs, Grenoble, France
| | - Henri Gröger
- EMBL Grenoble, 71 Avenue des Martyrs, Grenoble, France
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes (UGA), Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Centre National de la Recherche Scientifique (CNRS), Grenoble, France
| | - Luciano G Dolce
- EMBL Grenoble, 71 Avenue des Martyrs, Grenoble, France
- Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Université Grenoble-Alpes, Grenoble, France
| | - Cameron D Mackereth
- University of Bordeaux, INSERM, CNRS, ARNA Laboratory, U1212, UMR 5320, Bordeaux, France
| | - Ronald Micura
- Institute of Organic Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Jacek Jemielity
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
17
|
Khalid T, Malik A, Rasool N, Kanwal A, Nawaz H, Almas I. Cracking the code: the clinical and molecular impact of aminopyridines; a review (2019-2024). RSC Adv 2025; 15:688-711. [PMID: 39781020 PMCID: PMC11708541 DOI: 10.1039/d4ra07438f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Aminopyridines belong to a class of compounds that are monoamino and diamino derivatives of pyridine. They work primarily by blocking voltage-gated potassium channels in a dose-dependent manner. Essential heterocycles used extensively in synthetic, natural products, and medicinal chemistry are aminopyridine and its derivatives. A vast array of biological and pharmacological effects can result from the interaction of aminopyridine rings with different enzymes and receptors, due to their unique structural properties. Aminopyridine research is continually growing, and there are now greater expectations for how it may aid in the treatment of numerous disorders. This review article will serve as an innovative platform for researchers investigating aminopyridine compounds, intending thoroughly to examine both traditional and novel synthesis strategies in addition to investigating the various biological characteristics displayed by these adaptable heterocycles. We attempt to provide valuable insights that will contribute to further progress in the synthesis and utilization of aminopyridines in various fields.
Collapse
Affiliation(s)
- Tahira Khalid
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Ayesha Malik
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Nasir Rasool
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Aqsa Kanwal
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Hamna Nawaz
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| | - Iffat Almas
- Department of Chemistry, Government College University Faisalabad Faisalabad 38000 Pakistan
| |
Collapse
|
18
|
Hu S, Batool Z, Zheng X, Yang Y, Ullah A, Shen B. Exploration of innovative drug repurposing strategies for combating human protozoan diseases: Advances, challenges, and opportunities. J Pharm Anal 2025; 15:101084. [PMID: 39896318 PMCID: PMC11786068 DOI: 10.1016/j.jpha.2024.101084] [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: 05/09/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 02/04/2025] Open
Abstract
Protozoan infections (e.g., malaria, trypanosomiasis, and toxoplasmosis) pose a considerable global burden on public health and socioeconomic problems, leading to high rates of morbidity and mortality. Due to the limited arsenal of effective drugs for these diseases, which are associated with devastating side effects and escalating drug resistance, there is an urgent need for innovative antiprotozoal drugs. The emergence of drug repurposing offers a low-cost approach to discovering new therapies for protozoan diseases. In this review, we summarize recent advances in drug repurposing for various human protozoan diseases and explore cost-effective strategies to identify viable new treatments. We highlight the cross-applicability of repurposed drugs across diverse diseases and harness common chemical motifs to provide new insights into drug design, facilitating the discovery of new antiprotozoal drugs. Challenges and opportunities in the field are discussed, delineating novel directions for ongoing and future research.
Collapse
Affiliation(s)
- ShanShan Hu
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Zahra Batool
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Xin Zheng
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Yin Yang
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Amin Ullah
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Bairong Shen
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610213, China
| |
Collapse
|
19
|
Dos Santos Nascimento IJ, Santos MB, De Jesus Marinho WP, de Moura RO. Insights to Design New Drugs against Human African Trypanosomiasis Targeting Rhodesain using Covalent Docking, Molecular Dynamics Simulations, and MM-PBSA Calculations. Curr Comput Aided Drug Des 2025; 21:67-82. [PMID: 38310575 DOI: 10.2174/0115734099274797231205055827] [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: 09/10/2023] [Revised: 10/19/2023] [Accepted: 10/27/2023] [Indexed: 02/06/2024]
Abstract
BACKGROUND Neglected tropical diseases (NTDs) are parasitic and bacterial diseases that affect approximately 149 countries, mainly the poor population without basic sanitation. Among these, Human African Trypanosomiasis (HAT), known as sleeping sickness, shows alarming data, with treatment based on suramin and pentamidine in the initial phase and melarsoprol and eflornithine in the chronic phase. Thus, to discover new drugs, several studies point to rhodesain as a promising drug target due to the function of protein degradation and intracellular transport of proteins between the insect and host cells and is present in all cycle phases of the parasite. METHODS Here, based on the previous studies by Nascimento et al. (2021) [5], that show the main rhodesain inhibitors development in the last decade, molecular docking and dynamics were applied in these inhibitors datasets to reveal crucial information that can be into drug design. RESULTS Also, our findings using MD simulations and MM-PBSA calculations confirmed Gly19, Gly23, Gly65, Asp161, and Trp184, showing high binding energy (ΔGbind between -72.782 to -124.477 kJ.mol-1). In addition, Van der Waals interactions have a better contribution (-140,930 to -96,988 kJ.mol-1) than electrostatic forces (-43,270 to -6,854 kJ.mol-1), indicating Van der Waals interactions are the leading forces in forming and maintaining ligand-rhodesain complexes. Thus, conventional and covalent docking was employed and highlighted the presence of Michael acceptors in the ligands in a peptidomimetics scaffold, and interaction with Gly19, Gly23, Gly65, Asp161, and Trp184 is essential to the inhibiting activity. Furthermore, the Dynamic Cross-Correlation Maps (DCCM) show more correlated movements for all complexes than the free rhodesain and strong interactions in the regions of the aforementioned residues. Principal Component Analysis (PCA) demonstrates complex stability corroborating with RMSF and RMSD. CONCLUSION This study can provide valuable insights that can guide researchers worldwide to discover a new promising drug against HAT.
Collapse
Affiliation(s)
- Igor José Dos Santos Nascimento
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil
- Cesmac University Center, Pharmacy Departament, Maceió, Brazil
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| | - Mirelly Barbosa Santos
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| | - Washley Phyama De Jesus Marinho
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| | - Ricardo Olimpio de Moura
- Postgraduate Program in Pharmaceutical Sciences, State University of Paraíba, Campina Grande, 58429-500, Brazil
- Drug Development and Synthesis Laboratory, Department of Pharmacy, State University of Paraíba, Campina Grande, 58429-500, Brazil
| |
Collapse
|
20
|
Giraudo A, Bolchi C, Pallavicini M, Di Santo R, Costi R, Saccoliti F. Uncovering the Mechanism of Action of Antiprotozoal Agents: A Survey on Photoaffinity Labeling Strategy. Pharmaceuticals (Basel) 2024; 18:28. [PMID: 39861091 PMCID: PMC11768348 DOI: 10.3390/ph18010028] [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: 11/29/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Plasmodium, Leishmania, and Trypanosoma parasites are responsible for infectious diseases threatening millions of people worldwide. Despite more recent efforts devoted to the search for new antiprotozoal agents, efficacy, safety, and resistance issues still hinder the development of suited therapeutic options. The lack of robustly validated targets and the complexity of parasite's diseases have made phenotypic screening a preferential drug discovery strategy for the identification of new chemical entities. However, via this approach, no information on biological target(s) and mechanisms of action of compounds are provided. Among the target deconvolution strategies useful to fill this gap, photoaffinity labeling (PAL) has emerged as one of most suited to enable investigation in a complex cellular environment. More recently, PAL has been exploited to unravel the molecular basis of bioactive compounds' function in live parasites, allowing elucidation of the mechanism of action of both approved drugs and new chemical entities. Besides highlighting new potential drug targets, PAL can provide valuable information on efficacy and liabilities of small molecules at the molecular level, which could be exploited to greatly facilitate the rational optimization of compounds in terms of potency and safety. In this review, we will report the most recent studies that have leveraged PAL to disclose the biological targets and mechanism of action of phenotypically active compounds targeting kinetoplastid diseases (i.e., human African trypanosomiasis, leishmaniasis, and Chagas disease) and malaria. Moreover, we will comment on potential perspectives that this innovative approach can provide in aiding the discovery and development of new antiprotozoal drugs.
Collapse
Affiliation(s)
- Alessandro Giraudo
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Cristiano Bolchi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Marco Pallavicini
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Roberto Di Santo
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, “Sapienza” Università di Roma, p.le Aldo Moro 5, I-00185 Rome, Italy
| | - Roberta Costi
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, “Sapienza” Università di Roma, p.le Aldo Moro 5, I-00185 Rome, Italy
| | - Francesco Saccoliti
- Dipartimento di Scienze della Vita, della Salute e delle Professioni Sanitarie, Università degli Studi “Link Campus University”, Via del Casale di S. Pio V 44, I-00165 Rome, Italy
| |
Collapse
|
21
|
Liu LJ, Francisco KR, Sun YU, Serafim MSM, Amarasinghe DK, Teixeira TR, Lucero B, Kronenberger T, Elsayed W, Elwakeel H, Al-Hindy M, Almaliti J, Gerwick WH, O'Donoghue AJ, Caffrey CR. Carmaphycin B-Based Proteasome Inhibitors to Treat Human African Trypanosomiasis: Structure-Activity Relationship and In Vivo Efficacy. ACS Infect Dis 2024; 10:4182-4193. [PMID: 39589805 DOI: 10.1021/acsinfecdis.4c00441] [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] [Indexed: 11/28/2024]
Abstract
The proteasome is essential for eukaryotic cell proteostasis, and inhibitors of the 20S proteasome are progressing preclinically and clinically as antiparasitics. We screenedTrypanosoma brucei, the causative agent of human and animal African trypanosomiasis, in vitro with a set of 27 carmaphycin B analogs, irreversible epoxyketone inhibitors that were originally developed to inhibit thePlasmodium falciparum20S (Pf20S). The structure-activity relationship was distinct from that of the human c20S antitarget by the acceptance of d-amino acids at the P3 position of the peptidyl backbone to yield compounds with greatly decreased toxicity to human cells. For the three most selective compounds, binding to the Tb20S β5 catalytic subunit was confirmed by competition with a fluorescent activity-based probe. For one compound, J-80, with its P3 d-configuration, the differential binding to the parasite's β5 subunit was supported by both covalent and noncovalent docking analysis. Further, J-80 was equipotent against both Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense in vitro. In a mouse model of Stage 1 T. brucei infection, a single intraperitoneal (i.p.) dose of 40 mg/kg J-80 halted the growth of the parasite, and when given at 50 mg/kg i.p. twice daily for 5 days, parasitemia was decreased to below the detectable limit, with parasite recrudescence 48 h after the last dose. The in vivo proof of principle demonstrated by a potent, selective, and irreversible inhibitor of Tb20S reveals an alternative path to the development of kinetoplastid proteasome inhibitors that differs from the current focus on allosteric reversible inhibitors.
Collapse
Affiliation(s)
- Lawrence J Liu
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093, United States
| | - Karol R Francisco
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Yujie Uli Sun
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Mateus Sá Magalhães Serafim
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG Brazil, 31270901
| | - Dilini K Amarasinghe
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Thaiz R Teixeira
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Bobby Lucero
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093, United States
| | - Thales Kronenberger
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery (TüCAD2), Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
- Partner-site Tübingen, German Center for Infection Research (DZIF), Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - Waad Elsayed
- Department of Medical Parasitology, Faculty of Medicine, Ain Shams University, Abassia, Cairo 1181, Egypt
| | - Hala Elwakeel
- Department of Medical Parasitology, Faculty of Medicine, Ain Shams University, Abassia, Cairo 1181, Egypt
| | - Momen Al-Hindy
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jehad Almaliti
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Jordan, Amman 11942, Jordan
| | - William H Gerwick
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Anthony J O'Donoghue
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
22
|
Ombura FLO, Abd-Alla AM, Akutse KS, Runo S, Mireji PO, Bateta R, Otiwi JE, Ajene IJ, Khamis FM. Dual suppression of Glossina pallidipes using entomopathogenic fungal-based biopesticides and sterile insect technique. Front Microbiol 2024; 15:1472324. [PMID: 39717267 PMCID: PMC11663849 DOI: 10.3389/fmicb.2024.1472324] [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: 08/01/2024] [Accepted: 11/20/2024] [Indexed: 12/25/2024] Open
Abstract
Tsetse flies and trypanosomosis significantly impact bovine production and human health in sub-Saharan Africa, exacerbating underdevelopment, malnutrition, and poverty. Despite various control strategies, long-term success has been limited. This study evaluates the combined use of entomopathogenic fungi (EPF) and the sterile insect technique (SIT) to combat tsetse flies. Eleven EPF isolates were tested against teneral males of Glossina pallidipes, focusing on mortality rates, radial growth, and impacts on fly fitness. Temperature effects on conidial growth, sporulation, and spore yield of SIT-compatible/tolerant strains were also assessed. The fungal isolates significantly influenced mortality rates in both unirradiated and irradiated (SIT-treated) males (p < 0.0001). Metarhizium anisopliae strains ICIPE 20, ICIPE 32, ICIPE 41, ICIPE 62, ICIPE 78, and Beauveria bassiana ICIPE 603 showed higher SIT compatibility/tolerance with LT50 values of 11-30 days, compared to other more virulent isolates with LT50 values of 4-9 days. Temperature significantly affected the radial growth of SIT-compatible EPF strains (p < 0.0001), with M. anisopliae ICIPE 78 exhibiting the fastest conidia growth at 25°C. Spore yield varied significantly across temperatures (15-40°C), and the thermal range for conidia germination of SIT-compatible strains was 8.1-45.4°C, with an optimal range of 26.7-31.1°C. Moreover, infected unirradiated females and irradiated males (donors) successfully transmitted conidia to untreated flies (receivers) without significant differences in survival rates (p = 0.6438) and no observed sex dimorphism. Our findings highlight the potential of combining EPF and SIT as a novel dual approach that could effectively and synergistically suppress tsetse fly populations.
Collapse
Affiliation(s)
- Fidelis L. O. Ombura
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Adly M.M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Komivi S. Akutse
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Unit of Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Steven Runo
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Paul O. Mireji
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, Kikuyu, Kenya
- Centre for Geographic Medicine Research Coast, Kenya Medical Research Institute, Kilifi, Kenya
| | - Rosemary Bateta
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, Kikuyu, Kenya
| | - Joseck E. Otiwi
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Inusa J. Ajene
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Fathiya M. Khamis
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Department of Zoology and Entomology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
23
|
Alves DO, Geens R, da Silva Arruda HR, Jennen L, Corthaut S, Wuyts E, de Andrade GC, Prosdocimi F, Cordeiro Y, Pires JR, Vieira LR, de Oliveira GAP, Sterckx YGJ, Salmon D. Biophysical analysis of the membrane-proximal Venus Flytrap domain of ESAG4 receptor-like adenylate cyclase from Trypanosoma brucei. Mol Biochem Parasitol 2024; 260:111653. [PMID: 39447762 DOI: 10.1016/j.molbiopara.2024.111653] [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: 09/20/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
The protozoan parasite Trypanosoma brucei possesses a large family of transmembrane receptor-like adenylate cyclases (RACs), primarily located to the flagellar surface and involved in sensing of the extracellular environment. RACs exhibit a conserved topology characterized by a large N-terminal extracellular moiety harbouring two Venus Flytrap (VFT) bilobate structures separated from an intracellular catalytic domain by a single transmembrane helix. RAC activation, which typically occurs under mild acid stress, requires the dimerization of the intracellular catalytic domain. The occurrence of VFT domains in the RAC's extracellular moiety suggests their potential responsiveness to extracellular ligands in the absence of stress, although no such ligands have been identified so far. Herein we report the biophysical characterization of the membrane-proximal VFT2 domain of a bloodstream form-specific RAC called ESAG4, whose ectodomain 3D structure is completely unknown. The paper describes an AlphaFold2-based optimisation of the expression construct, enabling facile and high-yield recombinant production and purification of the target protein. Through an interdisciplinary approach combining various biophysical methods, we demonstrate that the optimised VFT2 domain obtained by recombination is properly folded and behaves as a monomer in solution. The latter suggests a ligand-binding capacity independent of dimerization, unlike typical mammalian VFT receptors, as guanylate cyclase. In silico VFT2 genomic analyses shows divergence among cyclase isoforms, hinting at ligand specificity. Taken together this improved procedure enabling facile and high-yield recombinant production and purification of the target protein could benefit researchers studying trypanosomal RAC VFT domains but also any trypanosome domain with poorly defined boundaries. Additionally, our findings support the stable monomeric VFT2 domain as a useful tool for future structural investigations and ligand screening.
Collapse
Affiliation(s)
- Desirée O Alves
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Av. Brigadeiro Trompowsky, Rio de Janeiro 21941-590, Brazil
| | - Rob Geens
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Wilrijk 2610, Belgium
| | - Hiam R da Silva Arruda
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Av. Brigadeiro Trompowsky, Rio de Janeiro 21941-590, Brazil
| | - Lisa Jennen
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Wilrijk 2610, Belgium
| | - Sam Corthaut
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Wilrijk 2610, Belgium
| | - Ellen Wuyts
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Wilrijk 2610, Belgium
| | - Guilherme Caldas de Andrade
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Av. Brigadeiro Trompowsky, Rio de Janeiro 21941-590, Brazil
| | - Francisco Prosdocimi
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Av. Brigadeiro Trompowsky, Rio de Janeiro 21941-590, Brazil
| | - Yraima Cordeiro
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, Rio de Janeiro 21941-902, Brazil
| | - José Ricardo Pires
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Av. Brigadeiro Trompowsky, Rio de Janeiro 21941-590, Brazil
| | - Larissa Rezende Vieira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Av. Brigadeiro Trompowsky, Rio de Janeiro 21941-590, Brazil
| | - Guilherme A P de Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Av. Brigadeiro Trompowsky, Rio de Janeiro 21941-590, Brazil
| | - Yann G-J Sterckx
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Wilrijk 2610, Belgium.
| | - Didier Salmon
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Av. Brigadeiro Trompowsky, Rio de Janeiro 21941-590, Brazil.
| |
Collapse
|
24
|
Snijders R, Shaw APM, Selby R, Tirados I, Bessell PR, Fukinsia A, Miaka E, Tediosi F, Hasker E, Antillon M. The cost of sleeping sickness vector control in Yasa Bonga, a health district in the Democratic Republic of the Congo. PLoS Negl Trop Dis 2024; 18:e0011959. [PMID: 39570979 DOI: 10.1371/journal.pntd.0011959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 12/23/2024] [Accepted: 11/01/2024] [Indexed: 12/25/2024] Open
Abstract
Gambiense human African trypanosomiasis (gHAT), a neglected tropical disease caused by a parasite transmitted by tsetse flies, once inflicted over 30,000 annual cases and resulted in an estimated half a million deaths in the late twentieth century. An international gHAT control program has reduced cases to under 1,000 annually, encouraging the World Health Organization to target the elimination of gHAT transmission by 2030. This requires adopting innovative disease control approaches in foci where transmission persists. Since the last decade, case detection and treatment, the mainstay of controlling the disease, is supplemented by vector control using Tiny Targets, small insecticide-treated screens, which attract and kill tsetse. The advantages of Tiny Targets lie in their relatively low cost, easy deployment, and effectiveness. The Democratic Republic of Congo (DRC), bearing 65% of the 799 gHAT cases reported globally in 2022, introduced Tiny Targets in 2015. This study estimates the annual cost of vector control using Tiny Targets in the health district of Yasa Bonga in the DRC and identifies the main cost drivers. Economic and financial costs, collected from the provider's perspective, were used to estimate the average cost of tsetse control expressed as cost (i) per target used, (ii) per target deployed, (iii) per linear kilometre of river controlled, and (iv) per square kilometre protected by vector control. Sensitivity analyses were conducted on key parameters for results robustness. The estimated annual economic cost for protecting an area of 1,925 km2 was 120,000 USD. This translates to 5.30 USD per target used each year, 11 USD per target deployed in the field, 573 USD per linear km treated, and 62 USD per km2 protected. These costs in the DRC are comparable to those in other countries. The study provides valuable information for practitioners and policymakers making rational, evidence-based decisions to control gHAT.
Collapse
Affiliation(s)
- Rian Snijders
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Alexandra P M Shaw
- University of Edinburgh, Edinburgh, United Kingdom
- AP Consultants, Walworth Enterprise Centre, Andover, United Kingdom
| | - Richard Selby
- Sightsavers, Haywards Heath, West Sussex, United Kingdom
| | - Inaki Tirados
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Alain Fukinsia
- Programme National de Lutte Contre la Trypanosomiase Humaine Africaine, Kinshasa, Democratic Republic of the Congo
| | - Erick Miaka
- Programme National de Lutte Contre la Trypanosomiase Humaine Africaine, Kinshasa, Democratic Republic of the Congo
| | - Fabrizio Tediosi
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Epco Hasker
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Marina Antillon
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
25
|
Xu N, Zhang X, Liu H, Xu Y, Lu H, Zhao L, He Y, Zhang M, Zhang J, Si G, Wang Z, Chen M, Cai Y, Zhang Y, Wang Q, Hao Y, Li Y, Zhou Z, Guo Y, Chang C, Liu M, Ma C, Wang Y, Fang L, Li S, Wang G, Liu Q, Liu W. Clinical and epidemiological investigation of human infection with zoonotic parasite Trypanosoma dionisii in China. J Infect 2024; 89:106290. [PMID: 39341404 DOI: 10.1016/j.jinf.2024.106290] [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: 08/13/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Trypanosomiasis continues to pose a global threat to human health, with human infection mainly caused by Trypanosoma brucei and Trypanosoma cruzi. METHODS We present a 30-year-old pregnant woman with persistent high fever from Shandong Province, China. High-throughput sequencing revealed the presence of Trypanosoma dionisii in blood. We conducted an analysis of the patient's clinical, epidemiological, and virological data. RESULTS The patients exhibited fever, shortness of breath, chest tightness, accompanied by change in liver function and inflammatory response. She made a full recovery without any long-term effects. T. dionisii was detected in blood collected 23 days after onset of illness. The 18S rRNA gene sequence showed close similarity to T. dionisii found in bats from Japan, while the gGAPDH gene was closely related to T. dionisii from bats in Mengyin County, Shandong Province. Phylogenetic analysis demonstrated the current T. dionisii belongs to clade B within its species group. Positive anti-Trypanosoma IgG antibody was detected from the patient on Day 23, 66 and 122 after disease onset, as well as the cord blood and serum from the newborn. Retrospective screening of wild small mammals captured from Shandong Province revealed a prevalence rate of 0.54% (7/1304) for T. dionisii; specifically among 0.81% (5/620) of Apodemus agrarius, and 0.46% (2/438) of Mus musculus. CONCLUSIONS The confirmation of human infection with T. dionisii underscores its potential as a zoonotic pathogen, while the widespread presence of this parasite in rodent and bat species emphasizes the emerging threat it poses to human health.
Collapse
Affiliation(s)
- Nannan Xu
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaoai Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100071, China
| | - Hui Liu
- Institute of Bacterial Disease, Jinan Center for Disease Control and Prevention, Jinan, Shandong 250021, China
| | - Yintao Xu
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Huixia Lu
- Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Jinan, Shandong 250012, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lianhui Zhao
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yishan He
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Meiqi Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100071, China
| | - Jingtao Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100071, China
| | - Guangqian Si
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100071, China
| | - Ziyi Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China
| | - Muxin Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China
| | - Yuchun Cai
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China
| | - Yi Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China
| | - Qiang Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China
| | - Yuwan Hao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China
| | - Yuanyuan Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China
| | - Zhengbin Zhou
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China
| | - Yunhai Guo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China
| | - Caiyun Chang
- Institute for Infectious Disease Control, Jinan Center for Disease Control and Prevention, Jinan, Shandong 250021, China
| | - Ming Liu
- Institute for Infectious Disease Control, Jinan Center for Disease Control and Prevention, Jinan, Shandong 250021, China
| | - Chuanmin Ma
- Institute of Bacterial Disease, Jinan Center for Disease Control and Prevention, Jinan, Shandong 250021, China
| | - Yongbin Wang
- Shandong Institute of Parasitic Disease, Shandong First Medical University (Shandong Academy of Medical Sciences), Jining, Shandong 272033, China
| | - Liqun Fang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100071, China
| | - Shizhu Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China.
| | - Gang Wang
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Qin Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Research Center for Tropical Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 20025, China.
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100071, China.
| |
Collapse
|
26
|
Tahir Aleem M, Munir F, Shakoor A, Ud Din Sindhu Z, Gao F. Advancement in the development of DNA vaccines against Trypanosoma brucei and future perspective. Int Immunopharmacol 2024; 140:112847. [PMID: 39088922 DOI: 10.1016/j.intimp.2024.112847] [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: 05/22/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 08/03/2024]
Abstract
Trypanosomes are the extracellular protozoan parasites that cause human African trypanosomiasis disease in humans and nagana disease in animals. Tsetse flies act as a vector for the transmission of the disease in African countries. Animals infected with these parasites become useless or workless, and if not treated, disease can be fatal. There are many side effects associated with old treatments and some of them result in death in 5% of cases. There is a major surface glycoprotein in the parasite known as variant surface glycoprotein. The immune system of the host develops antibodies against this antigen but due to antigenic variation, parasites evade the immune response. Currently, no vaccine is available that provides complete protection. In murine models, only partial protection was observed using certain antigens. In order to develop vaccines against trypanosomes, molecular biology and immunology tools have been used. Immunization is the sole method for the control of disease because the eradication of the vector from endemic areas is an impossible task. Genetic vaccines can carry multiple genes encoding different antigens of the same parasite or different parasites. DNA immunization induces the activation of both cellular immune response and humoral immune response along with the generation of memory. This review highlights the importance of DNA vaccines and advances in the development of DNA vaccines against T. brucei.
Collapse
Affiliation(s)
- Muhammad Tahir Aleem
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China; Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, College of Sciences and Health Professions, Cleveland State University, Cleveland, OH 44115, USA.
| | - Furqan Munir
- Department of Parasitology, Faculty of Veterinary Science, University of Agriculture, Faisalabad 38040, Pakistan
| | - Amna Shakoor
- Department of Anatomy, Faculty of Veterinary Science, University of Agriculture, Faisalabad 9, 38040, Pakistan
| | - Zia Ud Din Sindhu
- Department of Parasitology, Faculty of Veterinary Science, University of Agriculture, Faisalabad 38040, Pakistan
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
27
|
Soto-Sánchez J, Garza-Treviño G. Combination Therapy and Phytochemical-Loaded Nanosytems for the Treatment of Neglected Tropical Diseases. Pharmaceutics 2024; 16:1239. [PMID: 39458571 PMCID: PMC11510106 DOI: 10.3390/pharmaceutics16101239] [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: 08/06/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Neglected tropical diseases (NTDs), including leishmaniasis, trypanosomiasis, and schistosomiasis, impose a significant public health burden, especially in developing countries. Despite control efforts, treatment remains challenging due to drug resistance and lack of effective therapies. Objective: This study aimed to synthesize the current research on the combination therapy and phytochemical-loaded nanosystems, which have emerged as promising strategies to enhance treatment efficacy and safety. Methods/Results: In the present review, we conducted a systematic search of the literature and identified several phytochemicals that have been employed in this way, with the notable efficacy of reducing the parasite load in the liver and spleen in cases of visceral leishmaniasis, as well as lesion size in cutaneous leishmaniasis. Furthermore, they have a synergistic effect against Trypanosoma brucei rhodesiense rhodesain; reduce inflammation, parasitic load in the myocardium, cardiac hypertrophy, and IL-15 production in Chagas disease; and affect both mature and immature stages of Schistosoma mansoni, resulting in improved outcomes compared to the administration of phytochemicals alone or with conventional drugs. Moreover, the majority of the combinations studied demonstrated enhanced solubility, efficacy, and selectivity, as well as increased immune response and reduced cytotoxicity. Conclusions: These formulations appear to offer significant therapeutic benefits, although further research is required to validate their clinical efficacy in humans and their potential to improve treatment outcomes in affected populations.
Collapse
Affiliation(s)
- Jacqueline Soto-Sánchez
- Section for Postgraduate Studies and Research, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera #239, Fracc. La Escalera, Ticomán, Ciudad de México 07320, Mexico
| | - Gilberto Garza-Treviño
- Section for Postgraduate Studies and Research, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera #239, Fracc. La Escalera, Ticomán, Ciudad de México 07320, Mexico
| |
Collapse
|
28
|
Lopes A, Teixeira S, Santarém N, Greco A, Pagliaro A, Keminer O, Gul S, Cordeiro-da-Silva A, Carvalho MA. SAR Study of 4,8-Disubstituted Pyrimido[5,4- d]pyrimidines Exhibiting Antitrypanosomal and Antileishmanial Activity. ACS Med Chem Lett 2024; 15:1541-1548. [PMID: 39291018 PMCID: PMC11403736 DOI: 10.1021/acsmedchemlett.4c00277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
A set of new derivatives of 4,8-disubstituted pyrimido[5,4-d]pyrimidines were efficiently synthesized and in vitro evaluated against Trypanosoma brucei and Leishmania infantum promastigotes and intramacrophage amastigotes. The in vitro cytotoxicity was determined using the THP-1 cell line, and early in vitro ADME-Tox was carried out using in vitro assays for cytotoxicity (A549 and HEK293 cell lines) and CYP3A4 and hERG cardiotoxicity liabilities. All the new compounds were active against T. brucei (0.11 μM ≤ IC50 ≤ 8.72 μM; 1 ≤ selectivity index (SI) ≤ 877), but only eight were active against L. infantum promastigotes (0.20 μM ≤ IC50 ≤ 14.88 μM; 1 ≤ SI < 502) with three also active against L. infantum intramacrophage amastigotes (3.00 μM ≤ IC50 ≤ 8.51 μM). Compounds 4a, 4c, and 4n were identified as the hit compounds to further develop as antitrypanosomal and antileishmanial agents.
Collapse
Affiliation(s)
- André Lopes
- Centre of Chemistry of University of Minho (CQUM), Campus de Gualtar, Braga, Portugal and Departamento de Química, Escola de Ciências da Universidade do Minho, Braga 4710-057, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, University of Porto, Porto 4150-180, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto 4050-313, Portugal
| | - Sofia Teixeira
- Centre of Chemistry of University of Minho (CQUM), Campus de Gualtar, Braga, Portugal and Departamento de Química, Escola de Ciências da Universidade do Minho, Braga 4710-057, Portugal
| | - Nuno Santarém
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, University of Porto, Porto 4150-180, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto 4050-313, Portugal
| | - Alessandro Greco
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Hamburg 22525, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Hamburg 22525, Germany
| | - Angela Pagliaro
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Hamburg 22525, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Hamburg 22525, Germany
| | - Oliver Keminer
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Hamburg 22525, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Hamburg 22525, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Hamburg 22525, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Hamburg 22525, Germany
| | - Anabela Cordeiro-da-Silva
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, University of Porto, Porto 4150-180, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto 4050-313, Portugal
| | - Maria Alice Carvalho
- Centre of Chemistry of University of Minho (CQUM), Campus de Gualtar, Braga, Portugal and Departamento de Química, Escola de Ciências da Universidade do Minho, Braga 4710-057, Portugal
| |
Collapse
|
29
|
De Lira Silva NS, Schenkman S. Biogenesis of EVs in Trypanosomatids. CURRENT TOPICS IN MEMBRANES 2024; 94:49-83. [PMID: 39370213 DOI: 10.1016/bs.ctm.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Trypanosomes are protozoan parasites responsible for human diseases such as Chagas disease, African trypanosomiasis, and leishmaniasis. These organisms' growth in various environments and exhibit multiple morphological stages, while adapting their surface components. They acquire and release materials extensively to get nutrients and manage interactions with the extracellular environment. They acquire and utilize proteins, lipids, and carbohydrates for growth via using membrane transport and endocytosis. Endocytosis takes place through distinct membrane areas known as the flagellar pocket and cytostome, depending on the parasite species and its developmental stage. Some forms establish a complex endocytic system to either store or break down the absorbed materials. In contrast, membrane transport facilitates the uptake of small molecules like amino acids, carbohydrates, and iron via particular receptors on the plasma membrane. Concurrently, these parasites secrete various molecules such as proteins, enzymes, nucleic acids, and glycoconjugates either in soluble form or enclosed in extracellular vesicles, which significantly contribute to their parasitic behavior. These activities require exocytosis through a secretory pathway in certain membrane domains such as the flagellum, flagellar pocket, and plasma membrane, which are controlled at various developmental stages. The main features of the endocytic and exocytic mechanisms, as well as the organelles involved, are discussed in this chapter along with their connection to the formation of exosomes and extracellular vesicles in the Tritryp species.
Collapse
Affiliation(s)
- Nadjania Saraiva De Lira Silva
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Sergio Schenkman
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil; Antimicrobial Resistance Institute of São Paulo (Aries), São Paulo, Brazil.
| |
Collapse
|
30
|
Ferrins L, Diaz R, Cordon-Obras C, Rojas-Barros D, Quotadamo A, Oehme DP, Ceballos-Pérez G, Swaminathan U, Pérez-Moreno G, Bosch-Navarrete C, García-Hernández R, Gomez-Liñan C, Saura A, Ruiz-Perez LM, Gamarro F, Martinez-Martinez MS, Manzano P, González-Pacanowska D, Navarro M, Pollastri MP. Pharmacophore Identification and Structure-Activity Relationship Analysis of a Series of Substituted Azaindoles as Inhibitors of Trypanosoma brucei. J Med Chem 2024; 67:13985-14006. [PMID: 39136694 PMCID: PMC11345823 DOI: 10.1021/acs.jmedchem.4c00785] [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: 04/03/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
Human African trypanosomiasis is among the World Health Organization's designated neglected tropical diseases. Repurposing strategies are often employed in academic drug discovery programs due to financial limitations, and in this instance, we used human kinase inhibitor chemotypes to identify substituted 4-aminoazaindoles, exemplified by 1. Structure-activity and structure-property relationship analysis, informed by cheminformatics, identified 4s as a potent inhibitor of Trypanosoma brucei growth. While 4s appeared to be fast acting and cidal in the in vitro assays, it failed to cure a murine model of infection. Preliminary efforts to identify the potential mechanism of action of the series pointed to arginine kinase, though, as we demonstrate, this does not appear to be the sole target of our compounds. This comprehensive approach to drug discovery, encompassing cheminformatics, structure-potency and structure-property analysis, and pharmacophore identification, highlights our multipronged efforts to identify novel lead compounds for this deadly disease.
Collapse
Affiliation(s)
- Lori Ferrins
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Rosario Diaz
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Carlos Cordon-Obras
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Domingo Rojas-Barros
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Antonio Quotadamo
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
- Department
of Life Sciences, University of Modena and
Reggio Emilia, 41125 Modena, Italy
| | - Daniel P. Oehme
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Gloria Ceballos-Pérez
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Uma Swaminathan
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Guiomar Pérez-Moreno
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Cristina Bosch-Navarrete
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Raquel García-Hernández
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Claudia Gomez-Liñan
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Andreu Saura
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Luis Miguel Ruiz-Perez
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Francisco Gamarro
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | | | - Pilar Manzano
- Tres
Cantos R&D Center, GSK, Tres
Cantos 28760, Spain
| | - Dolores González-Pacanowska
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Miguel Navarro
- Instituto
de Parasitología y Biomedicina “López-Neyra”
Consejo Superior de Investigaciones Científicas (CSIC), Granada 18100, Spain
| | - Michael P. Pollastri
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| |
Collapse
|
31
|
Pazmiño-Betancourth M, Casas Gómez-Uribarri I, Mondragon-Shem K, Babayan SA, Baldini F, Rafuse Haines L. Advancing age grading techniques for Glossina morsitans morsitans, vectors of African trypanosomiasis, through mid-infrared spectroscopy and machine learning. Biol Methods Protoc 2024; 9:bpae058. [PMID: 39290986 PMCID: PMC11407438 DOI: 10.1093/biomethods/bpae058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Tsetse are the insects responsible for transmitting African trypanosomes, which cause sleeping sickness in humans and animal trypanosomiasis in wildlife and livestock. Knowing the age of these flies is important when assessing the effectiveness of vector control programs and modelling disease risk. Current methods to assess fly age are, however, labour-intensive, slow, and often inaccurate as skilled personnel are in short supply. Mid-infrared spectroscopy (MIRS), a fast and cost-effective tool to accurately estimate several biological traits of insects, offers a promising alternative. This is achieved by characterising the biochemical composition of the insect cuticle using infrared light coupled with machine-learning (ML) algorithms to estimate the traits of interest. We tested the performance of MIRS in estimating tsetse sex and age for the first-time using spectra obtained from their cuticle. We used 541 insectary-reared Glossina m. morsitans of two different age groups for males (5 and 7 weeks) and three age groups for females (3 days, 5 weeks, and 7 weeks). Spectra were collected from the head, thorax, and abdomen of each sample. ML models differentiated between male and female flies with a 96% accuracy and predicted the age group with 94% and 87% accuracy for males and females, respectively. The key infrared regions important for discriminating sex and age classification were characteristic of lipid and protein content. Our results support the use of MIRS as a rapid and accurate way to identify tsetse sex and age with minimal pre-processing. Further validation using wild-caught tsetse could pave the way for this technique to be implemented as a routine surveillance tool in vector control programmes.
Collapse
Affiliation(s)
- Mauro Pazmiño-Betancourth
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, G12 8QQ, Glasgow, United Kingdom
| | - Ivan Casas Gómez-Uribarri
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, G12 8QQ, Glasgow, United Kingdom
| | - Karina Mondragon-Shem
- Department of Vector Biology, Liverpool School of Tropical Medicine, L3 5QA, Liverpool, United Kingdom
| | - Simon A Babayan
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, G12 8QQ, Glasgow, United Kingdom
| | - Francesco Baldini
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, G12 8QQ, Glasgow, United Kingdom
- Environmental Health, and Ecological Sciences Department, Ifakara Health Institute, Morogoro, Ifakara, P.O. Box 53, United Republic of Tanzania
| | - Lee Rafuse Haines
- Department of Vector Biology, Liverpool School of Tropical Medicine, L3 5QA, Liverpool, United Kingdom
- Department of Biological Sciences, University of Notre Dame, 46556, Notre Dame, United States
| |
Collapse
|
32
|
Francesconi V, Rizzo M, Pozzi C, Tagliazucchi L, Konchie Simo CU, Saporito G, Landi G, Mangani S, Carbone A, Schenone S, Santarém N, Tavares J, Cordeiro-da-Silva A, Costi MP, Tonelli M. Identification of Innovative Folate Inhibitors Leveraging the Amino Dihydrotriazine Motif from Cycloguanil for Their Potential as Anti- Trypanosoma brucei Agents. ACS Infect Dis 2024; 10:2755-2774. [PMID: 38953453 PMCID: PMC11537224 DOI: 10.1021/acsinfecdis.4c00113] [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: 02/09/2024] [Revised: 05/06/2024] [Accepted: 06/11/2024] [Indexed: 07/04/2024]
Abstract
Folate enzymes, namely, dihydrofolate reductase (DHFR) and pteridine reductase (PTR1) are acknowledged targets for the development of antiparasitic agents against Trypanosomiasis and Leishmaniasis. Based on the amino dihydrotriazine motif of the drug Cycloguanil (Cyc), a known inhibitor of both folate enzymes, we have identified two novel series of inhibitors, the 2-amino triazino benzimidazoles (1) and 2-guanidino benzimidazoles (2), as their open ring analogues. Enzymatic screening was carried out against PTR1, DHFR, and thymidylate synthase (TS). The crystal structures of TbDHFR and TbPTR1 in complex with selected compounds experienced in both cases a substrate-like binding mode and allowed the rationalization of the main chemical features supporting the inhibitor ability to target folate enzymes. Biological evaluation of both series was performed against T. brucei and L. infantum and the toxicity against THP-1 human macrophages. Notably, the 5,6-dimethyl-2-guanidinobenzimidazole 2g resulted to be the most potent (Ki = 9 nM) and highly selective TbDHFR inhibitor, 6000-fold over TbPTR1 and 394-fold over hDHFR. The 5,6-dimethyl tricyclic analogue 1g, despite showing a lower potency and selectivity profile than 2g, shared a comparable antiparasitic activity against T. brucei in the low micromolar domain. The dichloro-substituted 2-guanidino benzimidazoles 2c and 2d revealed their potent and broad-spectrum antitrypanosomatid activity affecting the growth of T. brucei and L. infantum parasites. Therefore, both chemotypes could represent promising templates that could be valorized for further drug development.
Collapse
Affiliation(s)
- Valeria Francesconi
- Department
of Pharmacy, University of Genoa, viale Benedetto XV n.3, Genoa 16132, Italy
| | - Marco Rizzo
- Department
of Pharmacy, University of Genoa, viale Benedetto XV n.3, Genoa 16132, Italy
| | - Cecilia Pozzi
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, Siena 53100, Italy
- Consorzio
Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIMMP), Via Luigi Sacconi 6, Sesto Fiorentino (FI) 50019, Italy
| | - Lorenzo Tagliazucchi
- Department
of Life Science, University of Modena and
Reggio Emilia, via Campi 103, Modena 41125, Italy
- Doctorate
School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Via Campi 287, Modena 41125, Italy
| | - Claude U. Konchie Simo
- Department
of Life Science, University of Modena and
Reggio Emilia, via Campi 103, Modena 41125, Italy
| | - Giulia Saporito
- Department
of Life Science, University of Modena and
Reggio Emilia, via Campi 103, Modena 41125, Italy
| | - Giacomo Landi
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, Siena 53100, Italy
| | - Stefano Mangani
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, Siena 53100, Italy
| | - Anna Carbone
- Department
of Pharmacy, University of Genoa, viale Benedetto XV n.3, Genoa 16132, Italy
| | - Silvia Schenone
- Department
of Pharmacy, University of Genoa, viale Benedetto XV n.3, Genoa 16132, Italy
| | - Nuno Santarém
- i3S
- Institute
for Research and Innovation in Health, University
of Porto, Rua Alfredo
Allen, 208, Porto 4200-135, Portugal
| | - Joana Tavares
- i3S
- Institute
for Research and Innovation in Health, University
of Porto, Rua Alfredo
Allen, 208, Porto 4200-135, Portugal
| | - Anabela Cordeiro-da-Silva
- i3S
- Institute
for Research and Innovation in Health, University
of Porto, Rua Alfredo
Allen, 208, Porto 4200-135, Portugal
- Department
of Life Science, Faculty of Pharmacy, University
of Porto, Rua de Jorge
Viterbo Ferreira, 228, Porto 4050-313, Portugal
| | - Maria Paola Costi
- Department
of Life Science, University of Modena and
Reggio Emilia, via Campi 103, Modena 41125, Italy
| | - Michele Tonelli
- Department
of Pharmacy, University of Genoa, viale Benedetto XV n.3, Genoa 16132, Italy
| |
Collapse
|
33
|
Janse van Rensburg HD, Suganuma K, N'Da DD. In vitro trypanocidal activities and structure-activity relationships of ciprofloxacin analogs. Mol Divers 2024; 28:2667-2680. [PMID: 37481633 DOI: 10.1007/s11030-023-10704-9] [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: 03/14/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Tropical diseases, such as African trypanosomiasis, by their nature and prevalence lack the necessary urgency regarding drug development, despite the increasing need for novel, structurally diverse antitrypanosomal drugs, using different mechanisms of action that would improve drug efficacy and safety. Traditionally antibacterial agents, the fluoroquinolones, reportedly possess in vitro trypanocidal activities against Trypanosoma brucei organisms. During our research, the fluroquinolone, ciprofloxacin (1), and its analogs (2-24) were tested against bloodstream forms of T. brucei brucei, T. b. gambiense, T. b. rhodesiense, T. evansi, T. equiperdum, and T. congolense and Madin-Darby bovine kidney cells (cytotoxicity). Ciprofloxacin [CPX (1)] demonstrated selective trypanocidal activity against T. congolense (IC50 7.79 µM; SI 39.6), whereas the CPX derivatives (2-10) showed weak selective activity (25 < IC50 < 65 µM; 2 < SI < 4). Selectivity and activity of the CPX and 1,2,3-triazole (TZ) hybrids (11-24) were governed by their chemical functionality at C-3 (carboxylic acid, or 4-methylpiperazinyl amide) and their electronic effect (electron-donating or electron-withdrawing para-benzyl substituent), respectively. Trypanocidal hits in the micromolar range were identified against bloodstream forms of T. congolense [CPX (1); CPX amide derivatives 18: IC50 8.95 µM; SI 16.84; 22: IC50 5.42 µM; SI 25.2] and against T. brucei rhodesiense (CPX acid derivative 13: IC50 4.51 µM; SI 10.2), demonstrating more selectivity toward trypanosomes than mammalian cells. Hence, the trypanocidal hit compound 22 may be optimized by retaining the 4-methylpiperazine amide functional group (C-3) and the TZ moiety at position N-15 and introducing other electron-withdrawing ortho-, meta-, and/or para-substituents on the aryl ring in an effort to improve the pharmacokinetic properties and increase the trypanocidal activity.
Collapse
Affiliation(s)
| | - Keisuke Suganuma
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido, 080-8555, Japan.
| | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, 2520, South Africa
| |
Collapse
|
34
|
Tsague KJA, Bakwo Fils EM, Atagana JP, Mbeng DW, Palm L, Tchuinkam T, Schaer J. Molecular detection of trypanosomes of the Trypanosoma livingstonei species group in diverse bat species in Central Cameroon. Parasitol Res 2024; 123:280. [PMID: 39037445 PMCID: PMC11263222 DOI: 10.1007/s00436-024-08303-0] [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: 02/22/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Bats are hosts for diverse Trypanosoma species, including trypanosomes of the Trypanosoma cruzi clade. This clade is believed to have originated in Africa and diversified in many lineages worldwide. In several geographical areas, including Cameroon, no data about trypanosomes of bats has been collected yet. In this study, we investigated the diversity and phylogenetic relationships of trypanosomes of different bat species in the central region of Cameroon. Trypanosome infections were detected in six bat species of four bat families, namely Hipposideridae, Pteropodidae, Rhinolophidae, and Vespertilionidae, with an overall prevalence of 29% and the highest infection rate in hipposiderid bat species. All trypanosomes were identified as belonging to the Trypanosoma livingstonei species group with one clade that might represent an additional subspecies of T. livingstonei. Understanding the prevalence, distribution, and host range of parasites of this group contributes to our overall knowledge of the diversity and host specificity of trypanosome species that phylogenetically group at the base of the T. cruzi clade.
Collapse
Affiliation(s)
- K J A Tsague
- Laboratory of Biological Sciences, Faculty of Sciences of University of Maroua, Maroua, Cameroon
- Vector Borne Diseases Laboratory of the Research Unit for Biology and Applied Ecology (VBID-RUBAE), University of Dschang, Dschang, Cameroon
| | - E M Bakwo Fils
- Department of Environmental Sciences, Higher Institute of Agriculture, Forestry, Water and Environment (HIAFWE), University of Ebolowa, Ebolowa, Cameroon
| | - J P Atagana
- Department of Biological Science, Faculty of Science, University of Ngaoundere, Ngaoundere, Cameroon
| | - D W Mbeng
- Laboratory of Biological Sciences, Faculty of Sciences of University of Maroua, Maroua, Cameroon
| | - L Palm
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - T Tchuinkam
- Vector Borne Diseases Laboratory of the Research Unit for Biology and Applied Ecology (VBID-RUBAE), University of Dschang, Dschang, Cameroon.
| | - J Schaer
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.
- Department of Biology, Muni University, Arua, Uganda.
- Museum Für Naturkunde, Leibniz-Institute for Evolution and Biodiversity Science, Berlin, Germany.
- Department of Biological Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
35
|
Sinumvayo JP, Munezero PC, Tope AT, Adeyemo RO, Bale MI, Nyandwi JB, Haakuria VM, Mutesa L, Adedeji AA. Advancing Vaccinology Capacity: Education and Efforts in Vaccine Development and Manufacturing across Africa. Vaccines (Basel) 2024; 12:741. [PMID: 39066380 PMCID: PMC11281707 DOI: 10.3390/vaccines12070741] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
Africa, home to the world's second-largest population of approximately 1.3 billion, grapples with significant challenges in meeting its medical needs, particularly in accessing quality healthcare services and products. The continent faces a continuous onslaught of emerging infectious diseases, exacerbating the strain on its already fragile public health infrastructure. The COVID-19 crisis highlighted the urgency to build local vaccine production capacity and strengthen the health infrastructure in general. The risks associated with a heavy reliance on imported vaccines were exposed during the COVID-19 pandemic, necessitating the need to nurture and strengthen the local manufacturing of vaccines and therapeutic biologics. Various initiatives addressing training, manufacturing, and regulatory affairs are underway, and these require increasing dedicated and purposeful financial investment. Building vaccine manufacturing capacity requires substantial investment in training and infrastructure. This manuscript examines the current state of education in vaccinology and related sciences in Africa. It also provides an overview of the continent's efforts to address educational needs in vaccine development and manufacturing. Additionally, it evaluates the initiatives aimed at strengthening vaccine education and literacy, highlighting successful approaches and ongoing challenges. By assessing the progress made and identifying the remaining obstacles, this review offers insights into how Africa can enhance its vaccine manufacturing capacity to respond to vaccine-preventable disease challenges.
Collapse
Affiliation(s)
- Jean Paul Sinumvayo
- Department of Microbiology and Parasitology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye P.O. Box 117, Rwanda; (P.C.M.); (A.T.T.); (R.O.A.); (M.I.B.)
- East African Community, Regional Center of Excellence for Vaccines, Immunization and Health Supply Chain Management (EAC RCE-VIHSCM), Kigali P.O. Box 3286, Rwanda; (J.B.N.); (V.M.H.)
- Future of Medicine, Science, Technology and Innovation Research Group, School of Medicine and Pharmacy, University of Rwanda, Rwanda, Kigali P.O. Box 3286, Rwanda;
| | - Pierre Celestin Munezero
- Department of Microbiology and Parasitology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye P.O. Box 117, Rwanda; (P.C.M.); (A.T.T.); (R.O.A.); (M.I.B.)
- Future of Medicine, Science, Technology and Innovation Research Group, School of Medicine and Pharmacy, University of Rwanda, Rwanda, Kigali P.O. Box 3286, Rwanda;
| | - Adegboyega Taofeek Tope
- Department of Microbiology and Parasitology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye P.O. Box 117, Rwanda; (P.C.M.); (A.T.T.); (R.O.A.); (M.I.B.)
- Future of Medicine, Science, Technology and Innovation Research Group, School of Medicine and Pharmacy, University of Rwanda, Rwanda, Kigali P.O. Box 3286, Rwanda;
| | - Rasheed Omotayo Adeyemo
- Department of Microbiology and Parasitology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye P.O. Box 117, Rwanda; (P.C.M.); (A.T.T.); (R.O.A.); (M.I.B.)
- Future of Medicine, Science, Technology and Innovation Research Group, School of Medicine and Pharmacy, University of Rwanda, Rwanda, Kigali P.O. Box 3286, Rwanda;
| | - Muritala Issa Bale
- Department of Microbiology and Parasitology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye P.O. Box 117, Rwanda; (P.C.M.); (A.T.T.); (R.O.A.); (M.I.B.)
- Future of Medicine, Science, Technology and Innovation Research Group, School of Medicine and Pharmacy, University of Rwanda, Rwanda, Kigali P.O. Box 3286, Rwanda;
| | - Jean Baptiste Nyandwi
- East African Community, Regional Center of Excellence for Vaccines, Immunization and Health Supply Chain Management (EAC RCE-VIHSCM), Kigali P.O. Box 3286, Rwanda; (J.B.N.); (V.M.H.)
- Department of Pharmacology and Toxicology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye P.O. Box 117, Rwanda
| | - Vetjaera Mekupi Haakuria
- East African Community, Regional Center of Excellence for Vaccines, Immunization and Health Supply Chain Management (EAC RCE-VIHSCM), Kigali P.O. Box 3286, Rwanda; (J.B.N.); (V.M.H.)
| | - Leon Mutesa
- Department of Biochemistry, Molecular Biology and Genetics, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye P.O. Box 117, Rwanda;
- Center for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 4285, Rwanda
| | - Ahmed Adebowale Adedeji
- Future of Medicine, Science, Technology and Innovation Research Group, School of Medicine and Pharmacy, University of Rwanda, Rwanda, Kigali P.O. Box 3286, Rwanda;
- Department of Pharmacology and Toxicology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye P.O. Box 117, Rwanda
| |
Collapse
|
36
|
Mustière R, Dassonville-Klimpt A, Sonnet P. Aminopyridines in the development of drug candidates against protozoan neglected tropical diseases. Future Med Chem 2024; 16:1357-1373. [PMID: 39109436 PMCID: PMC11318709 DOI: 10.1080/17568919.2024.2359361] [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: 02/27/2024] [Accepted: 05/14/2024] [Indexed: 08/15/2024] Open
Abstract
Neglected tropical diseases (NTDs) pose a major threat in tropical zones for impoverished populations. Difficulty of access, adverse effects or low efficacy limit the use of current therapeutic options. Therefore, development of new drugs against NTDs is a necessity. Compounds containing an aminopyridine (AP) moiety are of great interest for the design of new anti-NTD drugs due to their intrinsic properties compared with their closest chemical structures. Currently, over 40 compounds with an AP moiety are on the market, but none is used against NTDs despite active research on APs. The aim of this review is to present the medicinal chemistry work carried out with these scaffolds, against protozoan NTDs: Trypanosoma cruzi, Trypanosoma brucei or Leishmania spp.
Collapse
Affiliation(s)
- Romain Mustière
- Université de Picardie-Jules-Verne, AGIR – Agents infectieux, RéSistance et chimiothérapie, UR 4294, UFR de pharmacie, 1, Rue des Louvels, F-80037 Amiens cedex 1, France
| | - Alexandra Dassonville-Klimpt
- Université de Picardie-Jules-Verne, AGIR – Agents infectieux, RéSistance et chimiothérapie, UR 4294, UFR de pharmacie, 1, Rue des Louvels, F-80037 Amiens cedex 1, France
| | - Pascal Sonnet
- Université de Picardie-Jules-Verne, AGIR – Agents infectieux, RéSistance et chimiothérapie, UR 4294, UFR de pharmacie, 1, Rue des Louvels, F-80037 Amiens cedex 1, France
| |
Collapse
|
37
|
Kennedy PGE. The evolving spectrum of human African trypanosomiasis. QJM 2024; 117:391-395. [PMID: 38065835 DOI: 10.1093/qjmed/hcad273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Indexed: 06/27/2024] Open
Abstract
Human African trypanosomiasis (HAT), or sleeping sickness, continues to be a major threat to human health in 36 countries throughout sub-Saharan Africa with up to 60 million people at risk. Over the last decade, there have been several advances in this area, some of which are discussed in this overview. Due to the concerted efforts of several bodies, including better identification and treatment of cases and improved tsetse fly vector control, the number of cases of HAT has declined dramatically. The clinical heterogeneity of HAT has also been increasingly recognized, and the disease, while usually fatal if untreated or inadequately treated, does not always have a uniformly fatal outcome. Improved methods of HAT diagnosis have now been developed including rapid diagnostic tests. Novel drug treatment of HAT has also been developed, notably nifurtimox-eflornithine combination therapy (NECT) for late-stage Trypanosoma brucei gambiense, oral fexinidazole for early and the early component of the late-stage of T.b. gambiense, and the new oral compounds of the oxaborole group, which have shown considerable promise in field trials. Advances in HAT neuropathogenesis have been steady, though largely incremental, with a particular focus on the role of the blood-brain barrier in parasite entry into the central nervous system and the relevant importance of both innate and adaptive immunity. While the World Health Organization goal of elimination of HAT as a public health problem by 2020 has probably been achieved, it remains to be seen whether the second more ambitious goal of interruption of transmission of HAT by 2030 will be attained.
Collapse
Affiliation(s)
- P G E Kennedy
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Wellcome Surgical Institute, Garscube Campus, Glasgow G61 1QH, UK
| |
Collapse
|
38
|
Corfu AI, Santarem N, Luelmo S, Mazza G, Greco A, Altomare A, Ferrario G, Nasta G, Keminer O, Aldini G, Tamborini L, Basilico N, Parapini S, Gul S, Cordeiro-da-Silva A, Conti P, Borsari C. Discovery of 1,3,4-Oxadiazole Derivatives as Broad-Spectrum Antiparasitic Agents. ACS Infect Dis 2024; 10:2222-2238. [PMID: 38717116 DOI: 10.1021/acsinfecdis.4c00181] [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] [Indexed: 06/15/2024]
Abstract
Vector-borne parasitic diseases (VBPDs) pose a significant threat to public health on a global scale. Collectively, Human African Trypanosomiasis (HAT), Leishmaniasis, and Malaria threaten millions of people, particularly in developing countries. Climate change might alter the transmission and spread of VBPDs, leading to a global burden of these diseases. Thus, novel agents are urgently needed to expand therapeutic options and limit the spread of drug-resistant parasites. Herein, we report the development of broad-spectrum antiparasitic agents by screening a known library of antileishmanial and antimalarial compounds toward Trypanosoma brucei (T. brucei) and identifying a 1,3,4-oxadiazole derivative (19) as anti-T. brucei hit with predicted blood-brain barrier permeability. Subsequently, extensive structure-activity-relationship studies around the lipophilic tail of 19 led to a potent antitrypanosomal and antimalarial compound (27), with moderate potency also toward Leishmania infantum (L. infantum) and Leishmania tropica. In addition, we discovered a pan-active antiparasitic molecule (24), showing low-micromolar IC50s toward T. brucei and Leishmania spp. promastigotes and amastigotes, and nanomolar IC50 against Plasmodium falciparum, together with high selectivity for the parasites over mammalian cells (THP-1). Early ADME-toxicity assays were used to assess the safety profile of the compounds. Overall, we characterized 24 and 27, bearing the 1,3,4-oxadiazole privileged scaffold, as broad-spectrum low-toxicity agents for the treatment of VBPDs. An alkyne-substituted chemical probe (30) was synthesized and will be utilized in proteomics experiments aimed at deconvoluting the mechanism of action in the T. brucei parasite.
Collapse
Affiliation(s)
- Alexandra Ioana Corfu
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Nuno Santarem
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sara Luelmo
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Gaia Mazza
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Alessandro Greco
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Alessandra Altomare
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Giulio Ferrario
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Giulia Nasta
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Oliver Keminer
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Lucia Tamborini
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Nicoletta Basilico
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Silvia Parapini
- Department of Biomedical Sciences for Health, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Anabela Cordeiro-da-Silva
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Paola Conti
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Chiara Borsari
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| |
Collapse
|
39
|
Hartman CB, Dube PS, Legoabe LJ, Van Pelt N, Matheeussen A, Caljon G, Beteck RM. Novel quinoline derivatives with broad-spectrum antiprotozoal activities. Arch Pharm (Weinheim) 2024; 357:e2300319. [PMID: 38396284 DOI: 10.1002/ardp.202300319] [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: 06/12/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024]
Abstract
Several quinoline derivatives incorporating arylnitro and aminochalcone moieties were synthesized and evaluated in vitro against a broad panel of trypanosomatid protozoan parasites responsible for sleeping sickness (Trypanosoma brucei rhodesiense), nagana (Trypanosoma brucei brucei), Chagas disease (Trypanosoma cruzi), and leishmaniasis (Leishmania infantum). Several of the compounds demonstrated significant antiprotozoal activity. Specifically, compounds 2c, 2d, and 4i displayed submicromolar activity against T. b. rhodesiense with half-maximal effective concentration (EC50) values of 0.68, 0.8, and 0.19 µM, respectively, and with a high selectivity relative to human lung fibroblasts and mouse primary macrophages (∼100-fold). Compounds 2d and 4i also showed considerable activity against T. b. brucei with EC50 values of 1.4 and 0.4 µM, respectively.
Collapse
Affiliation(s)
- Carla B Hartman
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Phelelisiwe S Dube
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Lesetja J Legoabe
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Natascha Van Pelt
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - An Matheeussen
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Richard M Beteck
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
40
|
Tsagmo JMN, Rotureau B, Calvo Alvarez E. Animal models of neglected parasitic diseases: In vivo multimodal imaging of experimental trypanosomatid infections. Methods Cell Biol 2024; 188:205-236. [PMID: 38880525 DOI: 10.1016/bs.mcb.2024.04.003] [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] [Indexed: 06/18/2024]
Abstract
African trypanosomiases and leishmaniases are significant neglected tropical diseases (NTDs) that affect millions globally, with severe health and socio-economic consequences, especially in endemic regions. Understanding the pathogenesis and dissemination of Trypanosoma brucei and Leishmania spp. parasites within their hosts is pivotal for the development of effective interventions. Whole-body bioluminescence and fluorescence imaging systems (BLI and FLI, respectively), are powerful tools to visualize and quantify the progression and distribution of these parasites in real-time within live animal models. By combining this technology with the engineering of stable T. brucei and Leishmania spp. strains expressing luciferase and/or fluorescent proteins, crucial aspects of the infection process including the parasites' homing, the infection dynamics, the tissue tropism, or the efficacy of experimental treatments and vaccines can be deeply investigated. This methodology allows for enhanced sensitivity and resolution, elucidating previously unrecognized infection niches and dynamics. Importantly, whole-body in vivo imaging is non-invasive, enabling for longitudinal studies during the course of an infection in the same animal, thereby aligning with the "3Rs" principle of animal research. Here, we detail a protocol for the generation of dual-reporter T. brucei and L. major, and their use to infect mice and follow the spatiotemporal dynamics of infection by in vivo imaging systems. Additionally, 3D micro-computed tomography (μCT) coupled to BLI in T. brucei-infected animals is applied to gain insights into the anatomical parasite distribution. This Chapter underscores the potential of these bioimaging modalities as indispensable tools in parasitology, paving the way for novel therapeutic strategies and deeper insights into host-parasite interactions.
Collapse
Affiliation(s)
- Jean Marc Ngoune Tsagmo
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, INSERM U1201, Department of Parasites and Insect Vectors, Institut Pasteur, Université Paris Cité, Paris, France
| | - Brice Rotureau
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, INSERM U1201, Department of Parasites and Insect Vectors, Institut Pasteur, Université Paris Cité, Paris, France; Parasitology Unit, Institut Pasteur of Guinea, Conakry, Guinea
| | | |
Collapse
|
41
|
Chen W, Zou R, Mei Y, Li J, Xuan Y, Cui B, Zou J, Wang J, Lin S, Zhang Z, Wang C. Structural insights into drug transport by an aquaglyceroporin. Nat Commun 2024; 15:3985. [PMID: 38734677 PMCID: PMC11088622 DOI: 10.1038/s41467-024-48445-4] [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: 10/29/2023] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Pentamidine and melarsoprol are primary drugs used to treat the lethal human sleeping sickness caused by the parasite Trypanosoma brucei. Cross-resistance to these two drugs has recently been linked to aquaglyceroporin 2 of the trypanosome (TbAQP2). TbAQP2 is the first member of the aquaporin family described as capable of drug transport; however, the underlying mechanism remains unclear. Here, we present cryo-electron microscopy structures of TbAQP2 bound to pentamidine or melarsoprol. Our structural studies, together with the molecular dynamic simulations, reveal the mechanisms shaping substrate specificity and drug permeation. Multiple amino acids in TbAQP2, near the extracellular entrance and inside the pore, create an expanded conducting tunnel, sterically and energetically allowing the permeation of pentamidine and melarsoprol. Our study elucidates the mechanism of drug transport by TbAQP2, providing valuable insights to inform the design of drugs against trypanosomiasis.
Collapse
Affiliation(s)
- Wanbiao Chen
- Center for Human Tissues and Organs Degeneration, Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 581055, China
| | - Rongfeng Zou
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Shenzhen, 518000, China
| | - Yi Mei
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China
| | - Jiawei Li
- Center for Human Tissues and Organs Degeneration, Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 581055, China
- Department of Geriatric Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, China
| | - Yumi Xuan
- Center for Human Tissues and Organs Degeneration, Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 581055, China
| | - Bing Cui
- Center for Human Tissues and Organs Degeneration, Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 581055, China
- School of Basic Medicine and Clinical Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Junjie Zou
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Shenzhen, 518000, China
| | - Juncheng Wang
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Shaoquan Lin
- Centre for Polymers in Medicine, Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 581055, China
| | - Zhe Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221004, China.
| | - Chongyuan Wang
- Center for Human Tissues and Organs Degeneration, Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 581055, China.
| |
Collapse
|
42
|
González-Montero MC, Andrés-Rodríguez J, García-Fernández N, Pérez-Pertejo Y, Reguera RM, Balaña-Fouce R, García-Estrada C. Targeting Trypanothione Metabolism in Trypanosomatids. Molecules 2024; 29:2214. [PMID: 38792079 PMCID: PMC11124245 DOI: 10.3390/molecules29102214] [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: 04/11/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Infectious diseases caused by trypanosomatids, including African trypanosomiasis (sleeping sickness), Chagas disease, and different forms of leishmaniasis, are Neglected Tropical Diseases affecting millions of people worldwide, mainly in vulnerable territories of tropical and subtropical areas. In general, current treatments against these diseases are old-fashioned, showing adverse effects and loss of efficacy due to misuse or overuse, thus leading to the emergence of resistance. For these reasons, searching for new antitrypanosomatid drugs has become an urgent necessity, and different metabolic pathways have been studied as potential drug targets against these parasites. Considering that trypanosomatids possess a unique redox pathway based on the trypanothione molecule absent in the mammalian host, the key enzymes involved in trypanothione metabolism, trypanothione reductase and trypanothione synthetase, have been studied in detail as druggable targets. In this review, we summarize some of the recent findings on the molecules inhibiting these two essential enzymes for Trypanosoma and Leishmania viability.
Collapse
Affiliation(s)
- María-Cristina González-Montero
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
| | - Julia Andrés-Rodríguez
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
| | - Nerea García-Fernández
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
| | - Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rosa M. Reguera
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
43
|
Gandra D, Mulama DH, Foureau DM, McKinney KQ, Kim E, Smith K, Haw J, Nagulapally A, Saulnier Sholler GL. DFMO inhibition of neuroblastoma tumorigenesis. Cancer Med 2024; 13:e7207. [PMID: 38686627 PMCID: PMC11058673 DOI: 10.1002/cam4.7207] [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: 04/01/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Most high-risk neuroblastoma patients who relapse succumb to disease despite the existing therapy. We recently reported increased event-free and overall survival in neuroblastoma patients receiving difluoromethylornithine (DFMO) during maintenance therapy. The effect of DFMO on cellular processes associated with neuroblastoma tumorigenesis needs further elucidation. Previous studies have shown cytotoxicity with IC50 values >5-15 mM, these doses are physiologically unattainable in patients, prompting further mechanistic studies at therapeutic doses. METHODS We characterized the effect of DFMO on cell viability, cell cycle, apoptosis, neurosphere formation, and protein expression in vitro using five established neuroblastoma cell lines (BE2C, CHLA-90, SHSY5Y, SMS-KCNR, and NGP) at clinically relevant doses of 0, 50, 100, 500, 1000, and 2500 μM. Limiting Dilution studies of tumor formation in murine models were performed. Statistical analysis was done using GraphPad and the level of significance set at p = 0.05. RESULTS There was not a significant loss of cell viability or gain of apoptotic activity in the in vitro assays (p > 0.05). DFMO treatment initiated G1 to S phase cell cycle arrest. There was a dose-dependent decrease in frequency and size of neurospheres and a dose-dependent increase in beta-galactosidase activity in all cell lines. Tumor formation was decreased in xenografts both with DFMO-pretreated cells and in mice treated with DFMO. CONCLUSION DFMO treatment is cytostatic at physiologically relevant doses and inhibits tumor initiation and progression in mice. This study suggests that DFMO, inhibits neuroblastoma by targeting cellular processes integral to neuroblastoma tumorigenesis at clinically relevant doses.
Collapse
Affiliation(s)
- Divya Gandra
- Department of PediatricsPenn State Health Children's HospitalHersheyPennsylvaniaUSA
| | - David H. Mulama
- Department of PediatricsLevine Children's HospitalCharlotteNorth CarolinaUSA
| | - David M. Foureau
- Department of MedicineLevine Cancer InstituteCharlotteNorth CarolinaUSA
| | | | - Elizabeth Kim
- Department of PediatricsLevine Children's HospitalCharlotteNorth CarolinaUSA
| | - Kaitlyn Smith
- Department of PediatricsLevine Children's HospitalCharlotteNorth CarolinaUSA
| | - Jason Haw
- Department of PediatricsLevine Children's HospitalCharlotteNorth CarolinaUSA
| | - Abhinav Nagulapally
- Department of PediatricsPenn State Health Children's HospitalHersheyPennsylvaniaUSA
| | | |
Collapse
|
44
|
Vahekeni N, Brillatz T, Rahmaty M, Cal M, Keller-Maerki S, Rocchetti R, Kaiser M, Sax S, Mattli K, Wolfram E, Marcourt L, Queiroz EF, Wolfender JL, Mäser P. Antiprotozoal Activity of Plants Used in the Management of Sleeping Sickness in Angola and Bioactivity-Guided Fractionation of Brasenia schreberi J.F.Gmel and Nymphaea lotus L. Active against T. b. rhodesiense. Molecules 2024; 29:1611. [PMID: 38611890 PMCID: PMC11013945 DOI: 10.3390/molecules29071611] [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: 01/15/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Folk medicine is widely used in Angola, even for human African trypanosomiasis (sleeping sickness) in spite of the fact that the reference treatment is available for free. Aiming to validate herbal remedies in use, we selected nine medicinal plants and assessed their antitrypanosomal activity. A total of 122 extracts were prepared using different plant parts and solvents. A total of 15 extracts from seven different plants exhibited in vitro activity (>70% at 20 µg/mL) against Trypanosoma brucei rhodesiense bloodstream forms. The dichloromethane extract of Nymphaea lotus (leaves and leaflets) and the ethanolic extract of Brasenia schreberi (leaves) had IC50 values ≤ 10 µg/mL. These two aquatic plants are of particular interest. They are being co-applied in the form of a decoction of leaves because they are considered by local healers as male and female of the same species, the ethnotaxon "longa dia simbi". Bioassay-guided fractionation led to the identification of eight active molecules: gallic acid (IC50 0.5 µg/mL), methyl gallate (IC50 1.1 µg/mL), 2,3,4,6-tetragalloyl-glucopyranoside, ethyl gallate (IC50 0.5 µg/mL), 1,2,3,4,6-pentagalloyl-β-glucopyranoside (IC50 20 µg/mL), gossypetin-7-O-β-glucopyranoside (IC50 5.5 µg/mL), and hypolaetin-7-O-glucoside (IC50 5.7 µg/mL) in B. schreberi, and 5-[(8Z,11Z,14Z)-heptadeca-8,11,14-trienyl] resorcinol (IC50 5.3 µg/mL) not described to date in N. lotus. Five of these active constituents were detected in the traditional preparation. This work provides the first evidence for the ethnomedicinal use of these plants in the management of sleeping sickness in Angola.
Collapse
Affiliation(s)
- Nina Vahekeni
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; (M.C.); (S.K.-M.); (R.R.); (M.K.); (S.S.); (P.M.)
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
| | - Théo Brillatz
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland; (T.B.); (L.M.); (E.F.Q.); (J.-L.W.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Marjan Rahmaty
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland; (T.B.); (L.M.); (E.F.Q.); (J.-L.W.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Monica Cal
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; (M.C.); (S.K.-M.); (R.R.); (M.K.); (S.S.); (P.M.)
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
| | - Sonja Keller-Maerki
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; (M.C.); (S.K.-M.); (R.R.); (M.K.); (S.S.); (P.M.)
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
| | - Romina Rocchetti
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; (M.C.); (S.K.-M.); (R.R.); (M.K.); (S.S.); (P.M.)
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
| | - Marcel Kaiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; (M.C.); (S.K.-M.); (R.R.); (M.K.); (S.S.); (P.M.)
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
| | - Sibylle Sax
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; (M.C.); (S.K.-M.); (R.R.); (M.K.); (S.S.); (P.M.)
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
| | - Kevin Mattli
- Phytopharmacy & Natural Products, Institute of Chemistry and Biotechnology, Zürich University of Applied Sciences (ZHAW), 8820 Wädenswil, Switzerland (E.W.)
| | - Evelyn Wolfram
- Phytopharmacy & Natural Products, Institute of Chemistry and Biotechnology, Zürich University of Applied Sciences (ZHAW), 8820 Wädenswil, Switzerland (E.W.)
| | - Laurence Marcourt
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland; (T.B.); (L.M.); (E.F.Q.); (J.-L.W.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Emerson Ferreira Queiroz
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland; (T.B.); (L.M.); (E.F.Q.); (J.-L.W.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland; (T.B.); (L.M.); (E.F.Q.); (J.-L.W.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Pascal Mäser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; (M.C.); (S.K.-M.); (R.R.); (M.K.); (S.S.); (P.M.)
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
| |
Collapse
|
45
|
Davis CN, Crump RE, Sutherland SA, Spencer SEF, Corbella A, Chansy S, Lebuki J, Miaka EM, Rock KS. Comparison of stochastic and deterministic models for gambiense sleeping sickness at different spatial scales: A health area analysis in the DRC. PLoS Comput Biol 2024; 20:e1011993. [PMID: 38557869 PMCID: PMC11008881 DOI: 10.1371/journal.pcbi.1011993] [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: 08/21/2023] [Revised: 04/11/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
The intensification of intervention activities against the fatal vector-borne disease gambiense human African trypanosomiasis (gHAT, sleeping sickness) in the last two decades has led to a large decline in the number of annually reported cases. However, while we move closer to achieving the ambitious target of elimination of transmission (EoT) to humans, pockets of infection remain, and it becomes increasingly important to quantitatively assess if different regions are on track for elimination, and where intervention efforts should be focused. We present a previously developed stochastic mathematical model for gHAT in the Democratic Republic of Congo (DRC) and show that this same formulation is able to capture the dynamics of gHAT observed at the health area level (approximately 10,000 people). This analysis was the first time any stochastic gHAT model has been fitted directly to case data and allows us to better quantify the uncertainty in our results. The analysis focuses on utilising a particle filter Markov chain Monte Carlo (MCMC) methodology to fit the model to the data from 16 health areas of Mosango health zone in Kwilu province as a case study. The spatial heterogeneity in cases is reflected in modelling results, where we predict that under the current intervention strategies, the health area of Kinzamba II, which has approximately one third of the health zone's cases, will have the latest expected year for EoT. We find that fitting the analogous deterministic version of the gHAT model using MCMC has substantially faster computation times than fitting the stochastic model using pMCMC, but produces virtually indistinguishable posterior parameterisation. This suggests that expanding health area fitting, to cover more of the DRC, should be done with deterministic fits for efficiency, but with stochastic projections used to capture both the parameter and stochastic variation in case reporting and elimination year estimations.
Collapse
Affiliation(s)
- Christopher N. Davis
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- School of Life Sciences, The University of Warwick, Coventry, United Kingdom
- Mathematics Institute, The University of Warwick, Coventry, United Kingdom
| | - Ronald E. Crump
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Mathematics Institute, The University of Warwick, Coventry, United Kingdom
| | - Samuel A. Sutherland
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Warwick Medical School, The University of Warwick, Coventry, United Kingdom
| | - Simon E. F. Spencer
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Department of Statistics, The University of Warwick, Coventry, United Kingdom
| | - Alice Corbella
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Department of Statistics, The University of Warwick, Coventry, United Kingdom
| | - Shampa Chansy
- Programme National de Lutte contre la Trypanosomiase Humaine Africaine (PNLTHA), Kinshasa, Democratic Republic of the Congo
| | - Junior Lebuki
- Programme National de Lutte contre la Trypanosomiase Humaine Africaine (PNLTHA), Kinshasa, Democratic Republic of the Congo
| | - Erick Mwamba Miaka
- Programme National de Lutte contre la Trypanosomiase Humaine Africaine (PNLTHA), Kinshasa, Democratic Republic of the Congo
| | - Kat S. Rock
- Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, The University of Warwick, Coventry, United Kingdom
- Mathematics Institute, The University of Warwick, Coventry, United Kingdom
| |
Collapse
|
46
|
Franco JR, Priotto G, Paone M, Cecchi G, Ebeja AK, Simarro PP, Sankara D, Metwally SBA, Argaw DD. The elimination of human African trypanosomiasis: Monitoring progress towards the 2021-2030 WHO road map targets. PLoS Negl Trop Dis 2024; 18:e0012111. [PMID: 38626188 PMCID: PMC11073784 DOI: 10.1371/journal.pntd.0012111] [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: 01/09/2024] [Revised: 05/06/2024] [Accepted: 03/26/2024] [Indexed: 04/18/2024] Open
Abstract
BACKGROUND Human African trypanosomiasis (HAT) is a neglected tropical disease that usually occurs in rural areas in sub-Saharan Africa. It caused devastating epidemics during the 20th century. Sustained, coordinated efforts by different stakeholders working with national sleeping sickness control programmes (NSSCPs) succeeded in controlling the disease and reducing the number of cases to historically low levels. In 2012, WHO targeted the elimination of the disease as a public health problem by 2020. This goal has been reached and a new ambitious target was stated in the WHO road map for NTDs 2021-2030 and endorsed by the 73rd World Health Assembly: the elimination of gambiense HAT transmission (i.e. reducing the number of reported cases to zero). The interruption of transmission was not considered as an achievable goal for rhodesiense HAT, as it would require vast veterinary interventions rather than actions at the public health level. METHODOLOGY/PRINCIPAL FINDINGS Data reported to WHO by NSSCPs were harmonized, verified, georeferenced and included in the atlas of HAT. A total of 802 cases were reported in 2021 and 837 in 2022. This is below the target for elimination as a public health problem at the global level (< 2000 HAT cases/year); 94% of the cases were caused by infection with T. b. gambiense. The areas reporting ≥ 1 HAT case/10 000 inhabitants/year in 2018-2022 cover a surface of 73 134 km2, with only 3013 km2 at very high or high risk. This represents a reduction of 90% from the baseline figure for 2000-2004, the target set for the elimination of HAT as a public health problem. For the surveillance of the disease, 4.5 million people were screened for gambiense HAT with serological tests in 2021-2022, 3.6 million through active screening and 0.9 million by passive screening. In 2021 and 2022 the elimination of HAT as a public health problem was validated in Benin, Uganda, Equatorial Guinea and Ghana for gambiense HAT and in Rwanda for rhodesiense HAT. To reach the next goal of elimination of transmission of gambiense HAT, countries have to report zero cases of human infection with T. b. gambiense for a period of at least 5 consecutive years. The criteria and procedures to verify elimination of transmission have been recently published by WHO. CONCLUSIONS/SIGNIFICANCE HAT elimination as a public health problem has been reached at global level, with seven countries already validated as having reached this goal. This achievement was made possible by the work of NSSCPs, supported by different public and private partners, and coordinated by WHO. The new challenging goal now is to reach zero cases by 2030. To reach this goal is crucial to maintain the engagement and support of donors and stakeholders and to keep the involvement and coordination of all partners. Along with the focus on elimination of transmission of gambiense HAT, it is important not to neglect rhodesiense HAT, which is targeted for elimination as a public health problem in the WHO road map for NTDs 2021-2030.
Collapse
Affiliation(s)
- Jose R. Franco
- World Health Organization, Global Neglected Tropical Diseases Programme, Prevention, Treatment and Care Unit, Geneva, Switzerland
| | - Gerardo Priotto
- World Health Organization, Global Neglected Tropical Diseases Programme, Prevention, Treatment and Care Unit, Geneva, Switzerland
| | - Massimo Paone
- Food and Agriculture Organization of the United Nations, Animal Production and Health Division, Rome, Italy
| | - Giuliano Cecchi
- Food and Agriculture Organization of the United Nations, Animal Production and Health Division, Rome, Italy
| | - Agustin Kadima Ebeja
- World Health Organization, Regional Office for Africa, Communicable Disease Unit, Brazzaville, Congo
| | - Pere P. Simarro
- Consultant, World Health Organization, Global Neglected Tropical Diseases Programme, Innovative and Intensified Disease Management Unit, Geneva, Switzerland
| | - Dieudonne Sankara
- World Health Organization, Global Neglected Tropical Diseases Programme, Prevention, Treatment and Care Unit, Geneva, Switzerland
| | - Samia B. A. Metwally
- Food and Agriculture Organization of the United Nations, Animal Production and Health Division, Rome, Italy
| | - Daniel Dagne Argaw
- World Health Organization, Global Neglected Tropical Diseases Programme, Prevention, Treatment and Care Unit, Geneva, Switzerland
| |
Collapse
|
47
|
Wendo JK, Mbaria JM, Nyariki JN, Isaac AO. Ginkgo biloba attenuated detrimental inflammatory and oxidative events due to Trypanosoma brucei rhodesiense in mice treated with melarsoprol. PLoS Negl Trop Dis 2024; 18:e0012103. [PMID: 38620045 PMCID: PMC11045140 DOI: 10.1371/journal.pntd.0012103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 04/25/2024] [Accepted: 03/25/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND The severe late stage Human African Trypanosomiasis (HAT) caused by Trypanosoma brucei rhodesiense (T.b.r) is characterized by damage to the blood brain barrier, severe brain inflammation, oxidative stress and organ damage. Melarsoprol (MelB) is currently the only treatment available for this disease. MelB use is limited by its lethal neurotoxicity due to post-treatment reactive encephalopathy. This study sought to assess the potential of Ginkgo biloba (GB), a potent anti-inflammatory and antioxidant, to protect the integrity of the blood brain barrier and ameliorate detrimental inflammatory and oxidative events due to T.b.r in mice treated with MelB. METHODOLOGY Group one constituted the control; group two was infected with T.b.r; group three was infected with T.b.r and treated with 2.2 mg/kg melarsoprol for 10 days; group four was infected with T.b.r and administered with GB 80 mg/kg for 30 days; group five was given GB 80mg/kg for two weeks before infection with T.b.r, and continued thereafter and group six was infected with T.b.r, administered with GB and treated with MelB. RESULTS Co-administration of MelB and GB improved the survival rate of infected mice. When administered separately, MelB and GB protected the integrity of the blood brain barrier and improved neurological function in infected mice. Furthermore, the administration of MelB and GB prevented T.b.r-induced microcytic hypochromic anaemia and thrombocytopenia, as well as T.b.r-driven downregulation of total WBCs. Glutathione analysis showed that co-administration of MelB and GB prevented T.b.r-induced oxidative stress in the brain, spleen, heart and lungs. Notably, GB averted peroxidation and oxidant damage by ameliorating T.b.r and MelB-driven elevation of malondialdehyde (MDA) in the brain, kidney and liver. In fact, the co-administered group for the liver, registered the lowest MDA levels for infected mice. T.b.r-driven elevation of serum TNF-α, IFN-γ, uric acid and urea was abrogated by MelB and GB. Co-administration of MelB and GB was most effective in stabilizing TNFα levels. GB attenuated T.b.r and MelB-driven up-regulation of nitrite. CONCLUSION Utilization of GB as an adjuvant therapy may ameliorate detrimental effects caused by T.b.r infection and MelB toxicity during late stage HAT.
Collapse
Affiliation(s)
- Janet Khatenje Wendo
- The University of Nairobi, Department of Public Health, Pharmacology and Toxicology, Kangemi (Nairobi), Kenya
- The Technical University of Kenya, Department of Pharmaceutical Sciences and Technology, Nairobi, Kenya
| | - James Mucunu Mbaria
- The University of Nairobi, Department of Public Health, Pharmacology and Toxicology, Kangemi (Nairobi), Kenya
| | - James Nyabuga Nyariki
- The Technical University of Kenya, Department of Biochemistry and Biotechnology, Nairobi, Kenya
| | - Alfred Orina Isaac
- The Technical University of Kenya, Department of Pharmaceutical Sciences and Technology, Nairobi, Kenya
| |
Collapse
|
48
|
Iribarren PA, Di Marzio LA, Berazategui MA, Saura A, Coria L, Cassataro J, Rojas F, Navarro M, Alvarez VE. Depolymerization of SUMO chains induces slender to stumpy differentiation in T. brucei bloodstream parasites. PLoS Pathog 2024; 20:e1012166. [PMID: 38635823 PMCID: PMC11060531 DOI: 10.1371/journal.ppat.1012166] [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: 01/17/2024] [Revised: 04/30/2024] [Accepted: 04/01/2024] [Indexed: 04/20/2024] Open
Abstract
Trypanosoma brucei are protozoan parasites that cause sleeping sickness in humans and nagana in cattle. Inside the mammalian host, a quorum sensing-like mechanism coordinates its differentiation from a slender replicative form into a quiescent stumpy form, limiting growth and activating metabolic pathways that are beneficial to the parasite in the insect host. The post-translational modification of proteins with the Small Ubiquitin-like MOdifier (SUMO) enables dynamic regulation of cellular metabolism. SUMO can be conjugated to its targets as a monomer but can also form oligomeric chains. Here, we have investigated the role of SUMO chains in T. brucei by abolishing the ability of SUMO to polymerize. We have found that parasites able to conjugate only SUMO monomers are primed for differentiation. This was demonstrated for monomorphic lines that are normally unable to produce stumpy forms in response to quorum sensing signaling in mice, and also for pleomorphic cell lines in which stumpy cells were observed at unusually low parasitemia levels. SUMO chain mutants showed a stumpy compatible transcriptional profile and better competence to differentiate into procyclics. Our study indicates that SUMO depolymerization may represent a coordinated signal triggered during stumpy activation program.
Collapse
Affiliation(s)
- Paula Ana Iribarren
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–IIBIO (UNSAM-CONICET), San Martin, Buenos Aires, Argentina
| | - Lucía Ayelén Di Marzio
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–IIBIO (UNSAM-CONICET), San Martin, Buenos Aires, Argentina
| | - María Agustina Berazategui
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–IIBIO (UNSAM-CONICET), San Martin, Buenos Aires, Argentina
| | - Andreu Saura
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC (IPBLN-CSIC), Granada, Spain
| | - Lorena Coria
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–IIBIO (UNSAM-CONICET), San Martin, Buenos Aires, Argentina
| | - Juliana Cassataro
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–IIBIO (UNSAM-CONICET), San Martin, Buenos Aires, Argentina
| | - Federico Rojas
- Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Miguel Navarro
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC (IPBLN-CSIC), Granada, Spain
| | - Vanina Eder Alvarez
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”–IIBIO (UNSAM-CONICET), San Martin, Buenos Aires, Argentina
| |
Collapse
|
49
|
Khartabil T, van Schaik RH, Haanstra JR, Koelewijn R, Russcher H, van Hellemond JJ. The fully automated Sysmex XN-31 hematology analyzer can detect bloodstream form Trypanosoma brucei. Diagn Microbiol Infect Dis 2024; 108:116193. [PMID: 38295683 DOI: 10.1016/j.diagmicrobio.2024.116193] [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: 10/26/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND For fully automated detection and quantification of Plasmodium parasites, Sysmex developed the XN-31 hemocytometer. This study investigated whether the XN-31 can also detect and quantify bloodstream form trypanosomes (trypomastigotes). METHODS Axenic cultures of Trypanosoma brucei brucei were used to prepare two dilution series of trypomastigotes in the whole blood of a healthy donor, which were subsequently examined by the XN-31 as well as by microscopic examination of thin and thick blood films. Trypomastigote intactness during the procedures was evaluated by microscopy. RESULTS The XN-31 hemocytometer detected trypomastigotes with a detection limit of 26 trypomastigotes/μL. Scattergram patterns of Trypanosoma and Plasmodium parasites were clearly distinct, but current interpretation settings do not allow the identification of trypomastigotes yet, and therefore, need future refinement. CONCLUSION Proof of concept was provided for an automated fluorescent flow cytometry method that can detect and quantify Plasmodium spp., as well as Trypanosoma brucei trypomastigotes.
Collapse
Affiliation(s)
- Tania Khartabil
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Ron Hn van Schaik
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Jurgen R Haanstra
- Systems Biology Lab, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Rob Koelewijn
- Department of Medical Microbiology & Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Henk Russcher
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Jaap J van Hellemond
- Department of Medical Microbiology & Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, the Netherlands.
| |
Collapse
|
50
|
Zahra M, Abrahamse H, George BP. Green nanotech paradigm for enhancing sesquiterpene lactone therapeutics in cancer. Biomed Pharmacother 2024; 173:116426. [PMID: 38471274 DOI: 10.1016/j.biopha.2024.116426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024] Open
Abstract
In the field of cancer therapy, sesquiterpene lactones (SLs) derived from diverse Dicoma species demonstrate noteworthy bioactivity. However, the translation of their full therapeutic potential into clinical applications encounters significant challenges, primarily related to solubility, bioavailability, and precise drug targeting. Despite these obstacles, our comprehensive review introduces an innovative paradigm shift that integrates the inherent therapeutic properties of SLs with the principles of green nanotechnology. To overcome issues of solubility, bioavailability, and targeted drug delivery, eco-friendly strategies are proposed for synthesizing nanocarriers. Green nanotechnology has emerged as a focal point in addressing environmental and health concerns linked to conventional treatments. This progressive approach of green nanotechnology holds promise for the development of safe and sustainable nanomaterials, particularly in the field of drug delivery. This groundbreaking methodology signifies a pioneering advancement in the creation of novel and effective anticancer therapeutics. It holds substantial potential for transforming cancer treatment and advancing the landscape of natural product research.
Collapse
Affiliation(s)
- Mehak Zahra
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 1711, Doornfontein 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 1711, Doornfontein 2028, South Africa
| | - Blassan P George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 1711, Doornfontein 2028, South Africa.
| |
Collapse
|