1
|
Gouegni EF, Yusuf AB, Habila AJ, Mamman M, Atawodi SE, Kato K, Inaoka DK, Hirayama K, Kita K, Shuaibu MN, Balogun EO. The extracellular region of Trypanosoma congolense Membrane Bound Acid Phosphatase induces partial protection in immunized BALB/c mice. Microb Pathog 2025; 204:107526. [PMID: 40185169 DOI: 10.1016/j.micpath.2025.107526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
African trypanosomiasis is a parasitic disease caused by African trypanosomes and there is no vaccine because the trypanosomes possess an intricate machinery for antigenic variation. Owing to the nonsusceptibility of trypanosomes' Membrane-Bound Acid Phosphatase (MBAP; EC 3.1.3.2) to antigenic variation and its central role in molecular trafficking, we hypothesized that MBAP is a promising target for vaccine development. Herein, a DNA vaccine was constructed using recombinant DNA technology to insert the gene of the extracellular portion (EP) of Trypanosoma congolense MBAP without its signal peptide (SP), into a Strep-tag/transin modified mammalian expression vector pVAX1. The construct (EP-SP/pVAX1) was transfected into HepG2 cells. Expression of the antigen (EP-SP) and the subcellular localization were confirmed by Immunofluorescence Assay. Thereafter, two independent vaccination trials with 100 μg EP-SP/pVAX1 were carried out in BALB/c mice. ELISA was used to check immunoglobulin G (IgG) and cytokines (IL-10 and IFN-ɣ) levels. The infection group were experimental infected with 104 T. congolense cells, and the potency of the vaccine was assessed by monitoring parasitemia and survival. There was a significant increase in circulating IgG (P = 0.048), IL-10 (P = 0.031) but no effect on IFN- ɣ (P = 1.001) in the animals. Low parasitaemia characterized by multiple waves with the intermittent clearing of parasites to no detectable levels, and extension of the lifespan by up to 45.45 % was observed with a complete survival of 20 % of vaccinated and infected mice. Together, the results showed that the vaccine construct was partially protective and has tremendous immunogenic potential, suggesting the needs for further studies.
Collapse
MESH Headings
- Animals
- Trypanosoma congolense/immunology
- Trypanosoma congolense/enzymology
- Trypanosoma congolense/genetics
- Mice, Inbred BALB C
- Mice
- Antibodies, Protozoan/blood
- Trypanosomiasis, African/prevention & control
- Trypanosomiasis, African/immunology
- Protozoan Vaccines/immunology
- Protozoan Vaccines/administration & dosage
- Protozoan Vaccines/genetics
- Acid Phosphatase/immunology
- Acid Phosphatase/genetics
- Humans
- Vaccines, DNA/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Immunoglobulin G/blood
- Antigens, Protozoan/immunology
- Antigens, Protozoan/genetics
- Female
- Interleukin-10/blood
- Hep G2 Cells
- Protozoan Proteins/immunology
- Protozoan Proteins/genetics
- Interferon-gamma/blood
- Disease Models, Animal
- Cytokines/blood
- Immunization
- Parasitemia/prevention & control
Collapse
Affiliation(s)
- Edwige F Gouegni
- Department of Biochemistry Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria; Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria; Africa Centre of Excellence for Mycotoxins and Food Safety, Federal University of Technology, Minna, 920211, Niger State, Nigeria
| | - Aminu B Yusuf
- Department of Biochemistry Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria; Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria
| | - Amaya J Habila
- Department of Biochemistry Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria; Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria
| | - Mohammed Mamman
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria; Department of Veterinary Pharmacology and Toxicology, Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria
| | - Sunday E Atawodi
- Department of Biochemistry, Federal University Lokoja, Lokoja, 260102, Kogi State, Nigeria
| | - Kentaro Kato
- Department of Eco-epidemiology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523, Japan; School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, 852-8523, Japan
| | - Daniel K Inaoka
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, 852-8523, Japan; Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523, Japan; Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kenji Hirayama
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, 852-8523, Japan
| | - Kiyoshi Kita
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, 852-8523, Japan; Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan; Department of Infection Biochemistry, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523, Japan
| | - Mohammed N Shuaibu
- Department of Biochemistry Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria; Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria
| | - Emmanuel O Balogun
- Department of Biochemistry Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria; Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, 810001, Kaduna State, Nigeria; Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan; Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC0657, La Jolla, CA, 92093, USA.
| |
Collapse
|
2
|
Kargbo A, Dafka S, Osman AM, Koua HK, Vieira RFC, Rocklöv J. Impact of climate change and variability on the occurrence and distribution of Trypanosoma vectors in The Gambia. Parasitol Res 2025; 124:29. [PMID: 40044953 PMCID: PMC11882701 DOI: 10.1007/s00436-025-08475-3] [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: 08/13/2024] [Accepted: 02/22/2025] [Indexed: 03/09/2025]
Abstract
Extreme weather events can lead to infectious disease outbreaks, especially those spread by hematophagous flies, and The Gambia is particularly vulnerable to climate change. To the best of our knowledge, no one has ever documented the relationship between climate variability and change and the distribution of the hematophagous flies belonging to the families Glossinidae, Tabanidae, and Stomoxyinae. This paper aims to study the association of temperature and humidity on the distribution of the above species and their families in The Gambia in the recent past and to provide predictions of species abundance and occurrence in the future. A line transect survey was carried out in all the administrative regions of The Gambia to study the prevalence of the flies. Generalized additive models were used to analyze the relationships between the distribution of the insects and their families and the variability in climate conditions in the recent past and in three different future periods. Regarding the recent past, our results show that temperature has significantly impacted the presence of Glossinidae and Tabanidae species, with maximum temperature being the most important factor. Relative humidity was also statistically significantly associated with Tabanidae species. None of the climate variables was found to be associated with the Tabanus par and Tabanus sufis. Minimum temperature and relative humidity were statistically significantly associated with Glossina morsitan submorsitan, while maximum temperature was statistically significantly associated with Atylotus agrestis and Stomoxys calcitrans. Only relative humidity was statistically significantly associated with the Glossina palpalis gambiense. As for the future projections, the results show that rising temperatures impacted the distribution of Tabanus species, Glossina species, and Stomoxys calcitrans in The Gambia. The distribution of Trypanosoma vectors in The Gambia is mostly influenced by maximum temperature. The research's conclusions gave climate and public health policymakers crucial information to take into account.
Collapse
Affiliation(s)
- Alpha Kargbo
- Department of Physical and Natural Sciences, University of the Gambia, P. O. Box 3530, Brikama Campus, Serrekunda, The Gambia.
- WASCAL-Graduate Research Program in Climate Change and Biodiversity, Universite Felix Houphouet-Boigny, BP V34, Abidjan, Côte d'Ivoire.
| | - Stella Dafka
- Heidelberg Institute of Global Health and Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany
| | - Aamir M Osman
- Graduate Program On Veterinary Sciences, Department of Veterinary Medicine, Universidade Federal Do Paraná, Curitiba, Paraná, Brazil
- Somali One Health Centre, Abrar University, Mogadishu, Somalia
- Department of Animal Health and Veterinary Services, Ministry of Livestock, Forestry, and Range, Mogadishu, Somalia
| | - Herve Kouakou Koua
- Laboratoire de Zoologie Et Biologie Animale, Université de Cocody, 22 BP 582 Abidjan 22, Abidjan, Côte d'Ivoire
| | - Rafael F C Vieira
- Center for Computational Intelligence to Predict Health and Environmental Risks (CIPHER), University of North Carolina at Charlotte, Charlotte, USA
- Department of Public Health Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Joacim Rocklöv
- Heidelberg Institute of Global Health and Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany.
- Department of Public Health and Clinical Medicine, Section of Sustainable Health, Umeå University, Umeå, Sweden.
| |
Collapse
|
3
|
Alzan HF, Mahmoud MS, Suarez CE. Current vaccines, experimental immunization trials, and new perspectives to control selected vector borne blood parasites of veterinary importance. Front Vet Sci 2024; 11:1484787. [PMID: 39606652 PMCID: PMC11602000 DOI: 10.3389/fvets.2024.1484787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Parasite infections transmitted by vectors such as ticks and blood-sucking arthropods pose a significant threat to both human and animal health worldwide and have a substantial economic impact, particularly in the context of worsening environmental conditions. These infections can manifest in a variety of symptoms, including fever, anemia, jaundice, enlarged spleen, neurological disorders, and lymphatic issues, and can have varying mortality rates. In this review, we will focus on the current state of available vaccines, vaccine research approaches, and trials for diseases caused by vector-borne blood parasites, such as Babesia, Theileria, Anaplasma, and Trypanosoma, in farm animals. Control measures for these infections primarily rely on vector control, parasiticidal drug treatments, and vaccinations for disease prevention. However, many of these approaches have limitations, such as environmental concerns associated with the use of parasiticides, acaricides, and insecticides. Additionally, while some vaccines for blood parasites are already available, they still have several drawbacks, including practicality issues, unsuitability in non-endemic areas, and concerns about spreading other infectious agents, particularly in the case of live vaccines. This article highlights recent efforts to develop vaccines for controlling blood parasites in animals. The focus is on vaccine development approaches that show promise, including those based on recombinant antigens, vectored vaccines, and live attenuated or genetically modified parasites. Despite intensive research, developing effective subunit vaccines against blood stage parasites remains a challenge. By learning from previous vaccine development efforts and using emerging technologies to define immune mechanisms of protection, appropriate adjuvants, and protective antigens, we can expand our toolkit for controlling these burdensome diseases.
Collapse
Affiliation(s)
- Heba F. Alzan
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
- Parasitology and Animal Diseases Department, Veterinary Research Institute, National Research Center, Giza, Egypt
| | - Mona S. Mahmoud
- Parasitology and Animal Diseases Department, Veterinary Research Institute, National Research Center, Giza, Egypt
| | - Carlos E. Suarez
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, WA, United States
| |
Collapse
|
4
|
Li J, Yang S, Wu Y, Wang R, Liu Y, Liu J, Ye Z, Tang R, Whiteway M, Lv Q, Yan L. Alternative Oxidase: From Molecule and Function to Future Inhibitors. ACS OMEGA 2024; 9:12478-12499. [PMID: 38524433 PMCID: PMC10955580 DOI: 10.1021/acsomega.3c09339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 03/26/2024]
Abstract
In the respiratory chain of the majority of aerobic organisms, the enzyme alternative oxidase (AOX) functions as the terminal oxidase and has important roles in maintaining metabolic and signaling homeostasis in mitochondria. AOX endows the respiratory system with flexibility in the coupling among the carbon metabolism pathway, electron transport chain (ETC) activity, and ATP turnover. AOX allows electrons to bypass the main cytochrome pathway to restrict the generation of reactive oxygen species (ROS). The inhibition of AOX leads to oxidative damage and contributes to the loss of adaptability and viability in some pathogenic organisms. Although AOXs have recently been identified in several organisms, crystal structures and major functions still need to be explored. Recent work on the trypanosome alternative oxidase has provided a crystal structure of an AOX protein, which contributes to the structure-activity relationship of the inhibitors of AOX. Here, we review the current knowledge on the development, structure, and properties of AOXs, as well as their roles and mechanisms in plants, animals, algae, protists, fungi, and bacteria, with a special emphasis on the development of AOX inhibitors, which will improve the understanding of respiratory regulation in many organisms and provide references for subsequent studies of AOX-targeted inhibitors.
Collapse
Affiliation(s)
- Jiye Li
- School
of Pharmacy, Naval Medical University, Shanghai 200433, China
- Institute
of Medicinal Biotechnology, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shiyun Yang
- School
of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Yujie Wu
- School
of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Ruina Wang
- School
of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Yu Liu
- School
of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Jiacun Liu
- School
of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Zi Ye
- School
of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Renjie Tang
- Beijing
South Medical District of Chinese PLA General Hospital, Beijing 100072, China
| | - Malcolm Whiteway
- Department
of Biology, Concordia University, Montreal, H4B 1R6 Quebec, Canada
| | - Quanzhen Lv
- School
of Pharmacy, Naval Medical University, Shanghai 200433, China
- Basic
Medicine Innovation Center for Fungal Infectious Diseases, (Naval Medical University), Ministry of Education, Shanghai 200433, China
- Key
Laboratory of Biosafety Defense (Naval Medical University), Ministry
of Education, Shanghai 200433, China
- Shanghai
Key Laboratory of Medical Biodefense, Shanghai 200433, China
| | - Lan Yan
- School
of Pharmacy, Naval Medical University, Shanghai 200433, China
- Basic
Medicine Innovation Center for Fungal Infectious Diseases, (Naval Medical University), Ministry of Education, Shanghai 200433, China
- Key
Laboratory of Biosafety Defense (Naval Medical University), Ministry
of Education, Shanghai 200433, China
- Shanghai
Key Laboratory of Medical Biodefense, Shanghai 200433, China
| |
Collapse
|
5
|
Danazumi AU, Ishmam IT, Idris S, Izert MA, Balogun EO, Górna MW. Targeted protein degradation might present a novel therapeutic approach in the fight against African trypanosomiasis. Eur J Pharm Sci 2023; 186:106451. [PMID: 37088149 PMCID: PMC11032742 DOI: 10.1016/j.ejps.2023.106451] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/11/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
African trypanosomiasis (AT) is a hemoparasitic disease caused by infection with African trypanosomes and it is prevalent in many sub-Saharan African countries, affecting both humans and domestic animals. The disease is transmitted mostly by haematophagous insects of the genus Glossina while taking blood meal, in the process spreading the parasites from an infected animal to an uninfected animal. The disease is fatal if untreated, and the available drugs are generally ineffective and resulting in toxicities. Therefore, it is still pertinent to explore novel methods and targets for drug discovery. Proteolysis-targeting chimeras (PROTACs) present a new strategy for development of therapeutic molecules that mimic cellular proteasomal-mediated protein degradation to target proteins involved in different disease types. PROTACs have been used to degrade proteins involved in various cancers, neurodegenerative diseases, and immune disorders with remarkable success. Here, we highlight the problems associated with the current treatments for AT, discuss the concept of PROTACs and associated targeted protein degradation (TPD) approaches, and provide some insights on the future potential for the use of these emerging technologies (PROTACs and TPD) for the development of new generation of anti-Trypanosoma drugs and the first "TrypPROTACs".
Collapse
Affiliation(s)
- Ammar Usman Danazumi
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland; Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland; Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | | | - Salisu Idris
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Matylda Anna Izert
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland
| | - Emmanuel Oluwadare Balogun
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria; African Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, Nigeria.
| | - Maria Wiktoria Górna
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
6
|
Ramirez Valdez K, Nzau B, Dorey-Robinson D, Jarman M, Nyagwange J, Schwartz JC, Freimanis G, Steyn AW, Warimwe GM, Morrison LJ, Mwangi W, Charleston B, Bonnet-Di Placido M, Hammond JA. A Customizable Suite of Methods to Sequence and Annotate Cattle Antibodies. Vaccines (Basel) 2023; 11:1099. [PMID: 37376488 PMCID: PMC10302312 DOI: 10.3390/vaccines11061099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Studying the antibody response to infection or vaccination is essential for developing more effective vaccines and therapeutics. Advances in high-throughput antibody sequencing technologies and immunoinformatic tools now allow the fast and comprehensive analysis of antibody repertoires at high resolution in any species. Here, we detail a flexible and customizable suite of methods from flow cytometry, single cell sorting, heavy and light chain amplification to antibody sequencing in cattle. These methods were used successfully, including adaptation to the 10x Genomics platform, to isolate native heavy-light chain pairs. When combined with the Ig-Sequence Multi-Species Annotation Tool, this suite represents a powerful toolkit for studying the cattle antibody response with high resolution and precision. Using three workflows, we processed 84, 96, and 8313 cattle B cells from which we sequenced 24, 31, and 4756 antibody heavy-light chain pairs, respectively. Each method has strengths and limitations in terms of the throughput, timeline, specialist equipment, and cost that are each discussed. Moreover, the principles outlined here can be applied to study antibody responses in other mammalian species.
Collapse
Affiliation(s)
| | - Benjamin Nzau
- The Pirbright Institute, Pirbright GU24 0NF, UK
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK
| | | | | | - James Nyagwange
- The Pirbright Institute, Pirbright GU24 0NF, UK
- KEMRI-Wellcome Trust Research Programme CGMRC, Kilifi P.O. Box 230-80108, Kenya
| | | | | | | | - George M. Warimwe
- KEMRI-Wellcome Trust Research Programme CGMRC, Kilifi P.O. Box 230-80108, Kenya
| | - Liam J. Morrison
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK
| | | | | | | | | |
Collapse
|
7
|
Alkhaldi AAM. Effects of Synthetic Ligustrazine-Based Chalcone Derivatives on Trypanosoma brucei brucei and Leishmania spp. Promastigotes. Molecules 2023; 28:4652. [PMID: 37375205 DOI: 10.3390/molecules28124652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Current medication therapy for leishmaniasis and trypanosomiasis remains a major challenge due to its limited efficacy, significant adverse effects, and inaccessibility. Consequently, locating affordable and effective medications is a pressing concern. Because of their easy-to-understand structure and high functionalization potential, chalcones are promising candidates for use as bioactive agents. Thirteen synthetic ligustrazine-containing chalcones were evaluated for their ability to inhibit the growth of leishmaniasis and trypanosomiasis in etiologic agents. The tetramethylpyrazine (TMP) analogue ligustrazine was chosen as the central moiety for the synthesis of these chalcone compounds. The most effective compound (EC50 = 2.59 µM) was the chalcone derivative 2c, which featured a pyrazin-2-yl amino on the ketone ring and a methyl substitution. Multiple actions were observed for certain derivatives, including 1c, 2a-c, 4b, and 5b, against all strains tested. Eflornithine served as a positive control, and three ligustrazine-based chalcone derivatives, including 1c, 2c, and 4b, had a higher relative potency. Compounds 1c and 2c are particularly efficacious; even more potent than the positive control, they are therefore promising candidates for the treatment of trypanosomiasis and leishmaniasis.
Collapse
|
8
|
Budania S, Dubey A, Singh A. Trypanosoma evansi RoTat 1.2 variant surface antigen mimotopes selected by panning of the random peptide phage-display library against monoclonal antibodies. J Mol Recognit 2022; 35:e2984. [PMID: 35869579 DOI: 10.1002/jmr.2984] [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: 06/20/2022] [Accepted: 07/13/2022] [Indexed: 12/24/2022]
Abstract
Mimotope peptides of native antigens are valuable for diverse applications such as diagnostics, therapeutics and modern vaccine design. Here, we report for the first time the selection and identification of peptide mimotopes of Trypanosoma evansi RoTat 1.2 variant surface glycoprotein (VSG) for their potential uses in surra diagnostics and multi-epitope vaccine research. First, we produced the mouse monoclonal antibodies (mAbs), designated as 2E11 (IgG1) and 1C2 (IgG1), against the antigens in T. evansi RoTat 1.2 lysates. We then used 2E11 mAb to immunoprecipitate the target antigen. The immunoprecipitated antigen was then identified to be the VSG by mass spectrometry. Both 2E11 and 1C2 mAbs reacted with the VSG in immunoblots. The surface plasmon resonance immunosensors developed with both the mAbs detected VSG in the parasite lysates as well as in the rodent sera. Further, the mAbs were biotinylated and used in three rounds of panning to select peptide mimotopes from the random peptide phage display library (PhD-12; New England Biolabs, USA). The phage clones selected against each mAb were amplified and tested by phage capture ELISA for specificity. The peptide coding regions of the selected phages were sequenced and the protein blast search of the deduced amino acid sequences was performed by accessing the non-redundant protein database at https://blast.ncbi.nlm.nih.gov/. The conformational B epitope prediction of the selected mimotope sequences was done by using 3D Pepitope algorithms accessed at: http://pepitope.tau.ac.il/. The potential applications of the selected mimotopes in surra diagnostics and research are being explored.
Collapse
Affiliation(s)
- Savita Budania
- Immunology Section, Department of Veterinary Microbiology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Abhishek Dubey
- Department of Biological Sciences, Indian Institute of Science Education & Research (IISER), Mohali, Punjab, India
| | - Ajit Singh
- Immunology Section, Department of Veterinary Microbiology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| |
Collapse
|
9
|
Danazumi AU, Iliyasu Gital S, Idris S, BS Dibba L, Balogun EO, Górna MW. Immunoinformatic design of a putative multi-epitope vaccine candidate against Trypanosoma brucei gambiense. Comput Struct Biotechnol J 2022; 20:5574-5585. [PMID: 36284708 PMCID: PMC9576565 DOI: 10.1016/j.csbj.2022.10.002] [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: 05/26/2022] [Revised: 09/13/2022] [Accepted: 10/02/2022] [Indexed: 11/28/2022] Open
Abstract
Human African trypanosomiasis (HAT) is a neglected tropical disease that is caused by flagellated parasites of the genus Trypanosoma. HAT imposes a significant socio-economic burden on many countries in sub-Saharan Africa and its control is hampered by several drawbacks ranging from the ineffectiveness of drugs, complex dosing regimens, drug resistance, and lack of a vaccine. Despite more than a century of research and investigations, the development of a vaccine to tackle HAT is still challenging due to the complex biology of the pathogens. Advancements in computational modeling coupled with the availability of an unprecedented amount of omics data from different organisms have allowed the design of new generation vaccines that offer better antigenicity and safety profile. One of such new generation approaches is a multi-epitope vaccine (MEV) designed from a collection of antigenic peptides. A MEV can stimulate both cellular and humoral immune responses as well as avoiding possible allergenic reactions. Herein, we take advantage of this approach to design a MEV from conserved hypothetical plasma membrane proteins of Trypanosoma brucei gambiense, the trypanosome subspecies that is responsible for the west and central African forms of HAT. The designed MEV is 402 amino acids long (41.5 kDa). It is predicted to be antigenic, non-toxic, to assume a stable 3D conformation, and to interact with a key immune receptor. In addition, immune simulation foresaw adequate immune stimulation by the putative antigen and a lasting memory. Therefore, the designed chimeric vaccine represents a potential candidate that could be used to target HAT.
Collapse
Affiliation(s)
- Ammar Usman Danazumi
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland,Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland,Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands,Corresponding authors at: Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland (A.U. Danazumi, M. W. Górna).
| | | | - Salisu Idris
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria,Department of Medical Laboratory Science, Kazaure School of Health Technology, Jigawa, Nigeria
| | - Lamin BS Dibba
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, Nigeria,Department of Physical and Natural Sciences, School of Arts and Sciences, University of the Gambia, Brikama Campus. P.O Box 3530, Serrekunda, the Gambia
| | - Emmanuel Oluwadare Balogun
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria,Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, Nigeria,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, CA 92093, USA,Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Maria Wiktoria Górna
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland,Corresponding authors at: Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland (A.U. Danazumi, M. W. Górna).
| |
Collapse
|
10
|
Romero-Ramirez A, Casas-Sánchez A, Autheman D, Duffy CW, Brandt C, Clare S, Harcourt K, André MR, de Almeida Castilho Neto KJG, Teixeira MMG, Machado RZ, Coombes J, Flynn RJ, Wright GJ, Jackson AP. Vivaxin genes encode highly immunogenic, non-variant antigens on the Trypanosoma vivax cell-surface. PLoS Negl Trop Dis 2022; 16:e0010791. [PMID: 36129968 PMCID: PMC9529106 DOI: 10.1371/journal.pntd.0010791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/03/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022] Open
Abstract
Trypanosoma vivax is a unicellular hemoparasite, and a principal cause of animal African trypanosomiasis (AAT), a vector-borne and potentially fatal livestock disease across sub-Saharan Africa. Previously, we identified diverse T. vivax-specific genes that were predicted to encode cell surface proteins. Here, we examine the immune responses of naturally and experimentally infected hosts to these unique parasite antigens, to identify immunogens that could become vaccine candidates. Immunoprofiling of host serum shows that one particular family (Fam34) elicits a consistent IgG antibody response. This gene family, which we now call Vivaxin, encodes at least 124 transmembrane glycoproteins that display quite distinct expression profiles and patterns of genetic variation. We focused on one gene (viv-β8) that encodes one particularly immunogenic vivaxin protein and which is highly expressed during infections but displays minimal polymorphism across the parasite population. Vaccination of mice with VIVβ8 adjuvanted with Quil-A elicits a strong, balanced immune response and delays parasite proliferation in some animals but, ultimately, it does not prevent disease. Although VIVβ8 is localized across the cell body and flagellar membrane, live immunostaining indicates that VIVβ8 is largely inaccessible to antibody in vivo. However, our phylogenetic analysis shows that vivaxin includes other antigens shown recently to induce immunity against T. vivax. Thus, the introduction of vivaxin represents an important advance in our understanding of the T. vivax cell surface. Besides being a source of proven and promising vaccine antigens, the gene family is clearly an important component of the parasite glycocalyx, with potential to influence host-parasite interactions. Animal African trypanosomiasis (AAT) is an important livestock disease throughout sub-Saharan Africa and beyond. AAT is caused by Trypanosoma vivax, among other species, a unicellular parasite that is spread by biting tsetse flies and multiplies in the bloodstream and other tissues, leading to often fatal neurological conditions if untreated. Although concerted drug treatment and vector eradication programmes have succeeded in controlling Human African trypanosomiasis, AAT continues to adversely affect animal health and impede efficient food production and economic development in many less-developed countries. In this study, we attempted to identify parasite surface proteins that stimulated the strongest immune responses in naturally infected animals, as the basis for a vaccine. We describe the discovery of a new, species-specific protein family in T. vivax, which we call vivaxin. We show that one vivaxin protein (VIVβ8) is surface expressed and retards parasite proliferation when used to immunize mice, but does not prevent infection. Nevertheless, we also reveal that vivaxin includes another protein previously shown to induce protective immunity (IFX/VIVβ1). Besides its great potential for novel approaches to AAT control, the vivaxin family is revealed as a significant component of the T. vivax cell surface and may have important, species-specific roles in host interactions.
Collapse
Affiliation(s)
- Alessandra Romero-Ramirez
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Aitor Casas-Sánchez
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Delphine Autheman
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Craig W. Duffy
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Cordelia Brandt
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Simon Clare
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Katherine Harcourt
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Marcos Rogério André
- Department of Pathology, Reproduction and One Health, Faculty of Agrarian and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, Sao Paulo, Brazil
| | - Kayo José Garcia de Almeida Castilho Neto
- Department of Pathology, Reproduction and One Health, Faculty of Agrarian and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, Sao Paulo, Brazil
| | - Marta M. G. Teixeira
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Rosangela Zacharias Machado
- Department of Pathology, Reproduction and One Health, Faculty of Agrarian and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, Sao Paulo, Brazil
| | - Janine Coombes
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- School of Pharmacy and Life Sciences, The Robert Gordon University, Aberdeen, United Kingdom
| | - Robin J. Flynn
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Waterford Institute of Technology, Waterford, Ireland
| | - Gavin J. Wright
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Andrew P. Jackson
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
11
|
Tauheed AM, Mamman M, Ahmed A, Suleiman MM, Balogun EO. Antitrypanosomal properties of Anogeissus leiocarpa extracts and their inhibitory effect on trypanosome alternative oxidase. PHYTOMEDICINE PLUS : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 2:100223. [PMID: 37378019 PMCID: PMC10295807 DOI: 10.1016/j.phyplu.2022.100223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Background African trypanosomiasis is a protozoan disease with huge socio-economic burden to sub-Saharan African exceeding US$4.6 annual loss. To mitigate the incidence of trypanosomal drug resistance, efforts are geared towards discovery of molecules, especially from natural products, with potential to inhibit important molecular target (trypanosome alternative oxidase, TAO) in trypanosomes that are critical to their survival. Method Crude methanol extract of Anogeissus leiocarpa was subjected to in vitro bioassay-guided antitrypanosomal assay to identify the most active extract with trypanocidal activity. The most active extract was run on a column chromatography yielding five fractions, F1-F5. The fractions were assayed for inhibitory effect on TAO. The most promising TAO inhibitor was subjected to antitrypanosomal evaluation by trypanosome count, drug incubation infectivity test (DIIT) and in vivo studies. Gas chromatography-mass spectrometry (GC-MS) was used to identify and quantify phytochemical constituents of the potential TAO-inhibiting fraction. Results Ethyl acetate extract (EtOAc) significantly (p<0.05) produced trypanocidal effect and was the most active extract. Of the five fractions, only F4 significantly (p<0.05) inhibited TAO compared to the control. F4 completely immobilised the trypanosomes up to 0.5 μg/μl, yielding an EC50 of 0.024 μg/μl compared to the 0.502 μg/μl of diminazene aceturate positive control group. The DIIT showed that F4 was significantly (p<0.05) potent up to 0.1 μg/μl. F4 significantly (p<0.05) suppressed parasite multiplication in systemic circulation of the treated rats and significantly (p<0.05) maintained high PCV when compared to the 5% DMSO group. Furthermore, F4 significantly (p<0.05) lowered serum concentrations of malondialdehyde. Phytoconstituents identified by the GC-MS include tetradecene; cetene; 3-(benzylthio) acrylic acid, methyl ester; 1-octadecene; 9-heptadecanone; hexadecanoic acid, methyl ester; dibutyl phthalate; eicosene; octadecenoic acid, methyl ester; oleic acid; 2-methyl-Z,Z-3,13-octadecadienol; 1-docosene; 3-phenylthiane, s-oxide; phenol, 3-methyl; phthalic acid, di(2-propylpentyl) ester and 1,4-benzenedicarboxylic acid, bis (2-ethylhexyl) ester. Conclusion F4 from EtOAc contains six carbohydrates (9.58%), two free fatty acids (6.48%), five fatty acid esters (27.73%), two aromatic compounds (50.63%) and one organosulphide (5.61%). It inhibited TAO and demonstrated antitrypanosomal effects.
Collapse
Affiliation(s)
- Abdullah M. Tauheed
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Mohammed Mamman
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Abubakar Ahmed
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Mohammed M. Suleiman
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Emmanuel O. Balogun
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology (ACENTDFB), Ahmadu Bello University, Zaria, Nigeria
| |
Collapse
|
12
|
Kargbo A, Ebiloma GU, Ibrahim YKE, Chechet GD, Jeng M, Balogun EO. Epizootiology and Molecular Identification of Trypanosome Species in Livestock Ruminants in the Gambia. Acta Parasitol 2022; 67:130-142. [PMID: 34164784 DOI: 10.1007/s11686-021-00442-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/14/2021] [Indexed: 11/25/2022]
Abstract
INTRODUCTION African Animal Trypanosomiasis (AAT) or nagana in animals, is caused by the blood-borne parasitic protozoa called trypanosomes, and is potentially fatal. It is estimated that Africa loses $4‒5 billion annually due to the death of livestock to nagana in the tsetse belt. PURPOSE Although The Gambia lies within this belt, there is scanty data regarding the epizootiology of nagana in The Gambia. Here, records of reported cases of nagana for the period 2010-2019 at the International Trypanotolerance Centre (ITC) in The Gambia were analyzed retrospectively. METHODS For insights into the current prevalence of AAT, blood samples of 384 cattle, 42 goats, and 59 sheep from the Central River Region (CRR) and Lower River Region (LRR) were analyzed microscopically for parasite identification. Furthermore, trypanosomes were characterized by polymerase chain reaction (PCR) using a panel of primers that identify trypanosomes to the level of the species and subspecies by targeting a portion of the internally transcribed spacer-one (ITS-1) of the ribosomal RNA. RESULTS The retrospective study indicates that Trypanosoma vivax (66%) and T. congolense (33.4%) were the predominant species. Based on the archive records of ITC, the villages Touba, Misera, and Sambel Kunda all in the CRR of the Gambia are the most burdened with AAT. Microscopic examination of blood samples from cattle showed a prevalence of 1.56%, whereas the PCR-based analysis gave a higher prevalence of 12.5%. The molecular analysis revealed the presence of T. vivax (3.65%), T. congolense kilifi (2.6%), T. b. brucei (1.3%), T. congolense savannah/forest (0.52%), T. b. gambiense (0.52%). Interestingly, 4.43% of mixed infections i.e. multiple trypanosome species in individual animals were recorded. In 18% of the mixed infection cases, T. godfreyi, T. simiae were coinfecting cattle alongside T. congolense. The molecular identification including the phylogenetic analysis implicated T. congolense as the most predominant trypanosome species infecting animals in The Gambia. CONCLUSION The incidence of nagana in The Gambia is documented and the prevalent trypanosomes identified to be T. vivax, different types of T. congolense, and T. brucei including the gambiense subspecie. Finally, nagana is less profound in sheep and goats compared to cattle, with seasonal and regional variations playing a significant role in the disease dynamics.
Collapse
Affiliation(s)
- Alpha Kargbo
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, P.M.B 1044, Zaria, Nigeria
- School of Arts and Sciences, University of The Gambia, MDI Road, Kanifing, P.O. Box 3530, Serrekunda, The Gambia
| | | | - Yakubu Kokori Enevene Ibrahim
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, P.M.B 1044, Zaria, Nigeria
- Department of Pharmaceutical Microbiology, Ahmadu Bello University, P.M.B 1044, Zaria, Kaduna State, Nigeria
| | - Gloria Dada Chechet
- Department of Biochemistry, Ahmadu Bello University, P.M.B 1044, Zaria, Kaduna State, Nigeria
| | - Momodou Jeng
- West Africa Livestock Innovation Center (WALIC/ITC), PMB 14, Banjul, The Gambia
| | - Emmanuel Oluwadare Balogun
- Department of Biochemistry, Ahmadu Bello University, P.M.B 1044, Zaria, Kaduna State, Nigeria.
- Department of Public Health and Epidemiology, Nigerian Institute of Medical Research, Yaba, P.M. B, Lagos, 2013, Nigeria.
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-Ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
13
|
Knowledge, Attitude, and Practice of Livestock Owners and Livestock Assistants towards African Trypanosomiasis Control in The Gambia. J Parasitol Res 2022; 2022:3379804. [PMID: 35111338 PMCID: PMC8803466 DOI: 10.1155/2022/3379804] [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: 09/09/2021] [Revised: 12/21/2021] [Accepted: 01/07/2022] [Indexed: 11/17/2022] Open
Abstract
Background In Africa, it has been estimated that 50 million cattle and 70 million sheep and goats are at risk of animal African trypanosomiasis, and three million cattle die annually. Methods This study was conducted in all the regions of The Gambia except Kombo Saint Mary Island (Banjul). Structured questionnaires were administered to 440 randomly selected livestock owners and 23 livestock assistants, and 7 focus group discussions were held for both livestock owners and livestock assistants. The data were analyzed mainly using descriptive statistics and content analysis methods. Results A total of 94.5% and 75% of livestock owners reported having seen tsetse and horse flies, respectively, while 100% of livestock assistants reported having seen tsetse flies. Forty-seven percent of the livestock owners indicated a positive attitude toward control measures, while 42% of them had no idea how to control tsetse flies. On the other hand, 57% of livestock assistants believe that tsetse and horse flies are the main reasons why AAT is still in their community. There was a statistically significant difference between all the respondents' characteristics and the practices done by livestock owners to prevent AAT vectors from biting their animals. Conclusion This study shows that trypanosomiasis is still a major problem for livestock health and production in The Gambia, and it requires disease and vector control.
Collapse
|
14
|
Adegboye O, Field MA, Kupz A, Pai S, Sharma D, Smout MJ, Wangchuk P, Wong Y, Loiseau C. Natural-Product-Based Solutions for Tropical Infectious Diseases. Clin Microbiol Rev 2021; 34:e0034820. [PMID: 34494873 PMCID: PMC8673330 DOI: 10.1128/cmr.00348-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
About half of the world's population and 80% of the world's biodiversity can be found in the tropics. Many diseases are specific to the tropics, with at least 41 diseases caused by endemic bacteria, viruses, parasites, and fungi. Such diseases are of increasing concern, as the geographic range of tropical diseases is expanding due to climate change, urbanization, change in agricultural practices, deforestation, and loss of biodiversity. While traditional medicines have been used for centuries in the treatment of tropical diseases, the active natural compounds within these medicines remain largely unknown. In this review, we describe infectious diseases specific to the tropics, including their causative pathogens, modes of transmission, recent major outbreaks, and geographic locations. We further review current treatments for these tropical diseases, carefully consider the biodiscovery potential of the tropical biome, and discuss a range of technologies being used for drug development from natural resources. We provide a list of natural products with antimicrobial activity, detailing the source organisms and their effectiveness as treatment. We discuss how technological advancements, such as next-generation sequencing, are driving high-throughput natural product screening pipelines to identify compounds with therapeutic properties. This review demonstrates the impact natural products from the vast tropical biome have in the treatment of tropical infectious diseases and how high-throughput technical capacity will accelerate this discovery process.
Collapse
Affiliation(s)
- Oyelola Adegboye
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
- World Health Organization Collaborating Center for Vector-Borne and Neglected Tropical Diseases, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Matt A. Field
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
- Garvin Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Andreas Kupz
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Saparna Pai
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Dileep Sharma
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- College of Medicine & Dentistry, James Cook University, Cairns, QLD, Australia
| | - Michael J. Smout
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Phurpa Wangchuk
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Yide Wong
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Claire Loiseau
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
15
|
Nguyen HTT, Guevarra RB, Magez S, Radwanska M. Single-cell transcriptome profiling and the use of AID deficient mice reveal that B cell activation combined with antibody class switch recombination and somatic hypermutation do not benefit the control of experimental trypanosomosis. PLoS Pathog 2021; 17:e1010026. [PMID: 34762705 PMCID: PMC8610246 DOI: 10.1371/journal.ppat.1010026] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 11/23/2021] [Accepted: 10/11/2021] [Indexed: 01/02/2023] Open
Abstract
Salivarian trypanosomes are extracellular protozoan parasites causing infections in a wide range of mammalian hosts, with Trypanosoma evansi having the widest geographic distribution, reaching territories far outside Africa and occasionally even Europe. Besides causing the animal diseases, T. evansi can cause atypical Human Trypanosomosis. The success of this parasite is attributed to its capacity to evade and disable the mammalian defense response. To unravel the latter, we applied here for the first time a scRNA-seq analysis on splenocytes from trypanosome infected mice, at two time points during infection, i.e. just after control of the first parasitemia peak (day 14) and a late chronic time point during infection (day 42). This analysis was combined with flow cytometry and ELISA, revealing that T. evansi induces prompt activation of splenic IgM+CD1d+ Marginal Zone and IgMIntIgD+ Follicular B cells, coinciding with an increase in plasma IgG2c Ab levels. Despite the absence of follicles, a rapid accumulation of Aicda+ GC-like B cells followed first parasitemia peak clearance, accompanied by the occurrence of Xbp1+ expressing CD138+ plasma B cells and Tbx21+ atypical CD11c+ memory B cells. Ablation of immature CD93+ bone marrow and Vpreb3+Ly6d+Ighm+ expressing transitional spleen B cells prevented mature peripheral B cell replenishment. Interestingly, AID-/- mice that lack the capacity to mount anti-parasite IgG responses, exhibited a superior defense level against T. evansi infections. Here, elevated natural IgMs were able to exert in vivo and in vitro trypanocidal activity. Hence, we conclude that in immune competent mice, trypanosomosis associated B cell activation and switched IgG production is rapidly induced by T. evansi, facilitating an escape from the detrimental natural IgM killing activity, and resulting in increased host susceptibility. This unique role of IgM and its anti-trypanosome activity are discussed in the context of the dilemma this causes for the future development of anti-trypanosome vaccines.
Collapse
Affiliation(s)
- Hang Thi Thu Nguyen
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robin B. Guevarra
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea
| | - Stefan Magez
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Magdalena Radwanska
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
16
|
Kasozi KI, Zirintunda G, Ssempijja F, Buyinza B, Alzahrani KJ, Matama K, Nakimbugwe HN, Alkazmi L, Onanyang D, Bogere P, Ochieng JJ, Islam S, Matovu W, Nalumenya DP, Batiha GES, Osuwat LO, Abdelhamid M, Shen T, Omadang L, Welburn SC. Epidemiology of Trypanosomiasis in Wildlife-Implications for Humans at the Wildlife Interface in Africa. Front Vet Sci 2021; 8:621699. [PMID: 34222391 PMCID: PMC8248802 DOI: 10.3389/fvets.2021.621699] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 05/05/2021] [Indexed: 12/18/2022] Open
Abstract
While both human and animal trypanosomiasis continue to present as major human and animal public health constraints globally, detailed analyses of trypanosome wildlife reservoir hosts remain sparse. African animal trypanosomiasis (AAT) affects both livestock and wildlife carrying a significant risk of spillover and cross-transmission of species and strains between populations. Increased human activity together with pressure on land resources is increasing wildlife–livestock–human infections. Increasing proximity between human settlements and grazing lands to wildlife reserves and game parks only serves to exacerbate zoonotic risk. Communities living and maintaining livestock on the fringes of wildlife-rich ecosystems require to have in place methods of vector control for prevention of AAT transmission and for the treatment of their livestock. Major Trypanosoma spp. include Trypanosoma brucei rhodesiense, Trypanosoma brucei gambiense, and Trypanosoma cruzi, pathogenic for humans, and Trypanosoma vivax, Trypanosoma congolense, Trypanosoma evansi, Trypanosoma brucei brucei, Trypanosoma dionisii, Trypanosoma thomasbancrofti, Trypanosma elephantis, Trypanosoma vegrandis, Trypanosoma copemani, Trypanosoma irwini, Trypanosoma copemani, Trypanosoma gilletti, Trypanosoma theileri, Trypanosoma godfreyi, Trypansoma simiae, and Trypanosoma (Megatrypanum) pestanai. Wildlife hosts for the trypansomatidae include subfamilies of Bovinae, Suidae, Pantherinae, Equidae, Alcephinae, Cercopithecinae, Crocodilinae, Pteropodidae, Peramelidae, Sigmodontidae, and Meliphagidae. Wildlife species are generally considered tolerant to trypanosome infection following centuries of coexistence of vectors and wildlife hosts. Tolerance is influenced by age, sex, species, and physiological condition and parasite challenge. Cyclic transmission through Glossina species occurs for T. congolense, T. simiae, T. vivax, T. brucei, and T. b. rhodesiense, T. b. gambiense, and within Reduviid bugs for T. cruzi. T. evansi is mechanically transmitted, and T. vixax is also commonly transmitted by biting flies including tsetse. Wildlife animal species serve as long-term reservoirs of infection, but the delicate acquired balance between trypanotolerance and trypanosome challenge can be disrupted by an increase in challenge and/or the introduction of new more virulent species into the ecosystem. There is a need to protect wildlife, animal, and human populations from the infectious consequences of encroachment to preserve and protect these populations. In this review, we explore the ecology and epidemiology of Trypanosoma spp. in wildlife.
Collapse
Affiliation(s)
- Keneth Iceland Kasozi
- Infection Medicine, Deanery of Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Scotland, United Kingdom.,School of Medicine, Kabale University, Kabale, Uganda
| | - Gerald Zirintunda
- Department of Animal Production and Management, Faculty of Agriculture and Animal Sciences, Busitema University Arapai Campus, Soroti, Uganda
| | - Fred Ssempijja
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Bridget Buyinza
- College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Kevin Matama
- School of Pharmacy, Kampala International University Western Campus, Bushenyi, Uganda
| | - Helen N Nakimbugwe
- Department of Animal Production and Management, Faculty of Agriculture and Animal Sciences, Busitema University Arapai Campus, Soroti, Uganda.,Department of Agriculture, Faculty of Vocational Studies, Kyambogo University, Kampala, Uganda
| | - Luay Alkazmi
- Biology Department, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - David Onanyang
- Department of Biology, Faculty of Science, Gulu University, Gulu, Uganda
| | - Paul Bogere
- Faculty of Agriculture and Environmental Science, Muni University, Arua, Uganda
| | - Juma John Ochieng
- Faculty of Biomedical Sciences, Kampala International University Western Campus, Bushenyi, Uganda
| | - Saher Islam
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
| | - Wycliff Matovu
- College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - David Paul Nalumenya
- College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | | | - Mahmoud Abdelhamid
- Department of Parasitology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| | - Tianren Shen
- Infection Medicine, Deanery of Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Scotland, United Kingdom.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Leonard Omadang
- Department of Animal Production and Management, Faculty of Agriculture and Animal Sciences, Busitema University Arapai Campus, Soroti, Uganda
| | - Susan Christina Welburn
- Infection Medicine, Deanery of Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Scotland, United Kingdom.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| |
Collapse
|
17
|
African Trypanosomosis Obliterates DTPa Vaccine-Induced Functional Memory So That Post-Treatment Bordetella pertussis Challenge Fails to Trigger a Protective Recall Response. Vaccines (Basel) 2021; 9:vaccines9060603. [PMID: 34200074 PMCID: PMC8230080 DOI: 10.3390/vaccines9060603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 11/17/2022] Open
Abstract
Salivarian trypanosomes are extracellular parasites causing anthroponotic and zoonotic infections. Anti-parasite vaccination is considered the only sustainable method for global trypanosomosis control. Unfortunately, no single field applicable vaccine solution has been successful so far. The active destruction of the host’s adaptive immune system by trypanosomes is believed to contribute to this problem. Here, we show that Trypanosome brucei brucei infection results in the lasting obliteration of immunological memory, including vaccine-induced memory against non-related pathogens. Using the well-established DTPa vaccine model in combination with a T. b. brucei infection and a diminazene diaceturate anti-parasite treatment scheme, our results demonstrate that while the latter ensured full recovery from the T. b. brucei infection, it failed to restore an efficacious anti-B. pertussis vaccine recall response. The DTPa vaccine failure coincided with a shift in the IgG1/IgG2a anti-B. pertussis antibody ratio in favor of IgG2a, and a striking impact on all of the spleen immune cell populations. Interestingly, an increased plasma IFNγ level in DTPa-vaccinated trypanosome-infected mice coincided with a temporary antibody-independent improvement in early-stage trypanosomosis control. In conclusion, our results are the first to show that trypanosome-inflicted immune damage is not restored by successful anti-parasite treatment.
Collapse
|
18
|
Salivarian Trypanosomes Have Adopted Intricate Host-Pathogen Interaction Mechanisms That Ensure Survival in Plain Sight of the Adaptive Immune System. Pathogens 2021; 10:pathogens10060679. [PMID: 34072674 PMCID: PMC8229994 DOI: 10.3390/pathogens10060679] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 12/21/2022] Open
Abstract
Salivarian trypanosomes are extracellular parasites affecting humans, livestock and game animals. Trypanosoma brucei rhodesiense and Trypanosoma brucei gambiense are human infective sub-species of T. brucei causing human African trypanosomiasis (HAT—sleeping sickness). The related T. b. brucei parasite lacks the resistance to survive in human serum, and only inflicts animal infections. Animal trypanosomiasis (AT) is not restricted to Africa, but is present on all continents. T. congolense and T. vivax are the most widespread pathogenic trypanosomes in sub-Saharan Africa. Through mechanical transmission, T. vivax has also been introduced into South America. T. evansi is a unique animal trypanosome that is found in vast territories around the world and can cause atypical human trypanosomiasis (aHT). All salivarian trypanosomes are well adapted to survival inside the host’s immune system. This is not a hostile environment for these parasites, but the place where they thrive. Here we provide an overview of the latest insights into the host-parasite interaction and the unique survival strategies that allow trypanosomes to outsmart the immune system. In addition, we review new developments in treatment and diagnosis as well as the issues that have hampered the development of field-applicable anti-trypanosome vaccines for the implementation of sustainable disease control.
Collapse
|
19
|
Memariani H, Memariani M. Melittin as a promising anti-protozoan peptide: current knowledge and future prospects. AMB Express 2021; 11:69. [PMID: 33983454 PMCID: PMC8119515 DOI: 10.1186/s13568-021-01229-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022] Open
Abstract
Protozoan diseases such as malaria, leishmaniasis, Chagas disease, and sleeping sickness still levy a heavy toll on human lives. Deplorably, only few classes of anti-protozoan drugs have thus far been developed. The problem is further compounded by their intrinsic toxicity, emergence of drug resistance, and the lack of licensed vaccines. Thus, there is a genuine exigency to develop novel anti-protozoan medications. Over the past years, melittin, the major constituent in the venom of European honeybee Apis mellifera, has gathered the attention of researchers due to its potential therapeutic applications. Insofar as we are aware, there has been no review pertinent to anti-protozoan properties of melittin. The present review outlines the current knowledge about anti-protozoan effects of melittin and its underlying mechanisms. The peptide has proven to be efficacious in killing different protozoan parasites such as Leishmania, Plasmodium, Toxoplasma, and Trypanosoma in vitro. Apart from direct membrane-disruptive activity, melittin is capable of destabilizing calcium homeostasis, reducing mitochondrial membrane potential, disorganizing kinetoplast DNA, instigating apoptotic cell death, and induction of autophagy in protozoan pathogens. Emerging evidence suggests that melittin is a promising candidate for future vaccine adjuvants. Transmission-blocking activity of melittin against vector-borne pathogens underscores its potential utility for both transgenic and paratransgenic manipulations. Nevertheless, future research should focus upon investigating anti-microbial activities of melittin, alone or in combination with the current anti-protozoan medications, against a far broader spectrum of protozoan parasites as well as pre-clinical testing of the peptide in animal models.
Collapse
|
20
|
VLP-Based Vaccines as a Suitable Technology to Target Trypanosomatid Diseases. Vaccines (Basel) 2021; 9:vaccines9030220. [PMID: 33807516 PMCID: PMC7998750 DOI: 10.3390/vaccines9030220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/25/2022] Open
Abstract
Research on vaccines against trypanosomatids, a family of protozoa that cause neglected tropical diseases, such as Chagas disease, leishmaniasis, and sleeping sickness, is a current need. Today, according to modern vaccinology, virus-like particle (VLP) technology is involved in many vaccines, including those undergoing studies related to COVID-19. The potential use of VLPs as vaccine adjuvants opens an opportunity for the use of protozoan antigens for the development of vaccines against diseases caused by Trypanosoma cruzi, Leishmania spp., and Trypanosoma brucei. In this context, it is important to consider the evasion mechanisms of these protozoa in the host and the antigens involved in the mechanisms of the parasite–host interaction. Thus, the immunostimulatory properties of VLPs can be part of an important strategy for the development and evaluation of new vaccines. This work aims to highlight the potential of VLPs as vaccine adjuvants for the development of immunity in complex diseases, specifically in the context of tropical diseases caused by trypanosomatids.
Collapse
|
21
|
Bouton J, Furquim d'Almeida A, Maes L, Caljon G, Van Calenbergh S, Hulpia F. Synthesis and evaluation of 3'-fluorinated 7-deazapurine nucleosides as antikinetoplastid agents. Eur J Med Chem 2021; 216:113290. [PMID: 33667845 DOI: 10.1016/j.ejmech.2021.113290] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/26/2021] [Accepted: 02/08/2021] [Indexed: 01/05/2023]
Abstract
Kinetoplastid parasites are the causative agents of neglected tropical diseases with an unmet medical need. These parasites are unable to synthesize the purine ring de novo, and therefore rely on purine salvage to meet their purine demand. Evaluating purine nucleoside analogs is therefore an attractive strategy to identify antikinetoplastid agents. Several anti-Trypanosoma cruzi and anti-Trypanosoma brucei 7-deazapurine nucleosides were previously discovered, with the removal of the 3'-hydroxyl group resulting in a significant boost in activity. In this work we therefore decided to assess the effect of the introduction of a 3'-fluoro substituent in 7-deazapurine nucleosides on the anti-kinetoplastid activities. Hence, we synthesized two series of 3'-deoxy-3'-fluororibofuranosyl and 3'-deoxy-3'-fluoroxylofuranosyl nucleosides comprising 7-deazaadenine and -hypoxanthine bases and assayed these for antiparasitic activity. Several analogs with potent activity against T. cruzi and T. brucei were discovered, indicating that a fluorine atom in the 3'-position is a promising modification for the discovery of antiparasitic nucleosides.
Collapse
Affiliation(s)
- Jakob Bouton
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000, Gent, Belgium
| | - Arno Furquim d'Almeida
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000, Gent, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610, Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610, Wilrijk, Belgium
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000, Gent, Belgium.
| | - Fabian Hulpia
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000, Gent, Belgium
| |
Collapse
|
22
|
Vehicular Livestock Mobility in West Africa: Seasonal Traffic Flows of Cattle, Sheep, and Goats across Bamako. SUSTAINABILITY 2020. [DOI: 10.3390/su13010171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mali is a major livestock producing country in West Africa. However, in recent years, the sector has faced multiple challenges like farmer-herder conflicts, overuse of grazing and water resources, and the effects of climate change. Meanwhile, traditional livestock systems are becoming less important given the increased availability of vehicular transport for regional and international animal trade as well as new opportunities for more specialised urban and peri-urban production systems. To assess the role of Mali’s capital city Bamako for livestock consumption and trade, this study examined the scale of livestock movement in the city, comprising influxes, outfluxes, and transits. To this end, flows of cattle, small ruminants, and livestock feed were recorded through road surveys covering three different seasons. The results showed the role of Bamako as a major trade hub in the distribution of ruminants. While traffic of cattle and sheep was regional, movement of goats was smaller and more localised. Religious festivals were an important driver of livestock traffic, both in terms of the numbers of livestock and the distance covered. This study highlights the role of cities for livestock mobility and contributes to a better understanding of challenges related to urban livestock production and trade and its requirements for better livestock management.
Collapse
|
23
|
Tauheed AM, Mamman M, Ahmed A, Suleiman MM, Balogun EO. In vitro and in vivo antitrypanosomal efficacy of combination therapy of Anogeissus leiocarpus, Khaya senegalensis and potash. JOURNAL OF ETHNOPHARMACOLOGY 2020; 258:112805. [PMID: 32243988 DOI: 10.1016/j.jep.2020.112805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/11/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pastoralists in Nigeria mix barks of Anogeissus leiocarpus (AL) Khaya senegalensis (KS) and potash (Pt) to treat animal African trypanosomosis. AIM To evaluate antitrypanosomal potential of A. leiocarpus, K. senegalensis and potash for insights into the traditional claim of antitrypanosomal combination therapy (ATCT). MATERIALS AND METHODS Fifty microliter each of six different concentrations of AL, KS, Pt, AL + KS, AL + KS + Pt and diminazene aceturate (DA, positive control) was incubated with 50 μL of parasite-laden blood containing 108Trypanosoma congolense cells in a 96-well microtitre plate. Negative control wells were devoid of the extracts and drug but supplemented with phosphate-buffered saline (PBS). Efficacy of treatment was observed at 1 h interval for complete immobilisation or reduced motility of the parasites. Each incubated mixture was inoculated into mouse at the point of complete immobilisation of parasite motility or at the end of 6-h observation period for concentrations that did not immobilise the parasites completely. For in vivo assessment, thirty-five parasitaemic rats were randomly allocated into seven groups of 5 rats each. Each rat in groups I-V was treated with 500 mg/kg of AL, KS, Pt, AL + KS and AL + KS + Pt, respectively, for 7 days. Rats in groups VI and VII were treated with diminazene aceturate 3.5 mg/kg once and PBS 2 mL/kg (7 days), which served as positive and negative controls, respectively. Daily monitoring of parasitaemia through the tail vein, packed cell volume and malondialdehyde were used to assess efficacy of the treatments. RESULTS The AL + KS + Pt group significantly (p < 0.05) and dose-dependently reduced parasite motility and completely immobilized the parasites at 10, 5 and 2.5 μg/μL with an IC50 of 9.1×10-4 µg/µL. All the mice with conditions that produced complete cessation of parasite motility did not develop parasitaemia within one month of observation. The AL + KS group significantly (p < 0.05) lowered the level of parasitaemia and MDA, and significantly (p < 0.05) maintained higher PCV than PBS group. CONCLUSION The combination of A. leiocarpus and K. senegalensis showed better antitrypanosomal effects than single drug treatment and offers prospects for ATCT. Our findings support ethnopharmacological use of combined barks of A. leiocarpus and K. senegalensis by pastoralist in the treatment of animal African trypanosomosis in Nigeria.
Collapse
Affiliation(s)
- Abdullah M Tauheed
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, Kaduna State, Nigeria.
| | - Mohammed Mamman
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Abubakar Ahmed
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Mohammed M Suleiman
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, Kaduna State, Nigeria; College of Agriculture and Animal Science, Mando, Ahmadu Bello University, Kaduna State, Nigeria
| | - Emmanuel O Balogun
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria; School of Pharmaceutical Sciences, University of California San Diego, United States of America
| |
Collapse
|
24
|
Ashour DS, Othman AA. Parasite-bacteria interrelationship. Parasitol Res 2020; 119:3145-3164. [PMID: 32748037 DOI: 10.1007/s00436-020-06804-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/06/2020] [Indexed: 12/18/2022]
Abstract
Parasites and bacteria have co-evolved with humankind, and they interact all the time in a myriad of ways. For example, some bacterial infections result from parasite-dwelling bacteria as in the case of Salmonella infection during schistosomiasis. Other bacteria synergize with parasites in the evolution of human disease as in the case of the interplay between Wolbachia endosymbiont bacteria and filarial nematodes as well as the interaction between Gram-negative bacteria and Schistosoma haematobium in the pathogenesis of urinary bladder cancer. Moreover, secondary bacterial infections may complicate several parasitic diseases such as visceral leishmaniasis and malaria, due to immunosuppression of the host during parasitic infections. Also, bacteria may colonize the parasitic lesions; for example, hydatid cysts and skin lesions of ectoparasites. Remarkably, some parasitic helminths and arthropods exhibit antibacterial activity usually by the release of specific antimicrobial products. Lastly, some parasite-bacteria interactions are induced as when using probiotic bacteria to modulate the outcome of a variety of parasitic infections. In sum, parasite-bacteria interactions involve intricate processes that never cease to intrigue the researchers. However, understanding and exploiting these interactions could have prophylactic and curative potential for infections by both types of pathogens.
Collapse
Affiliation(s)
- Dalia S Ashour
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, 31527, Egypt.
| | - Ahmad A Othman
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
25
|
Inflammation following trypanosome infection and persistence in the skin. Curr Opin Immunol 2020; 66:65-73. [PMID: 32446136 DOI: 10.1016/j.coi.2020.04.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023]
Abstract
Human African trypanosomes rely for their transmission on tsetse flies (Glossina sp.) that inoculate parasites into the skin during blood feeding. The absence of a protective vaccine, limited knowledge about the infection immunology, and the existence of asymptomatic carriers sustaining transmission are major outstanding challenges towards elimination. All these relate to the skin where (i) parasites persist and transmit to tsetse flies and (ii) a successful vaccination strategy should ideally be effective. Host immune processes and parasite strategies that underlie early infection and skin tropism are essential aspects to comprehend the transmission-success of trypanosomes and the failure in vaccine development. Recent insights into the early infection establishment may pave the way to novel strategies aimed at blocking transmission.
Collapse
|
26
|
Design of an Epitope-Based Vaccine Ensemble for Animal Trypanosomiasis by Computational Methods. Vaccines (Basel) 2020; 8:vaccines8010130. [PMID: 32188062 PMCID: PMC7157688 DOI: 10.3390/vaccines8010130] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/29/2020] [Accepted: 03/13/2020] [Indexed: 12/22/2022] Open
Abstract
African animal trypanosomiasis is caused by vector-transmitted parasites of the genus Trypanosoma. T. congolense and T. brucei brucei are predominant in Africa; T. evansi and T. vivax in America and Asia. They have in common an extracellular lifestyle and livestock tropism, which provokes huge economic losses in regions where vectors are endemic. There are licensed drugs to treat the infections, but adherence to treatment is poor and appearance of resistances common. Therefore, the availability of a prophylactic vaccine would represent a major breakthrough towards the management and control of the disease. Selection of the most appropriate antigens for its development is a bottleneck step, especially considering the limited resources allocated. Herein we propose a vaccine strategy based on multiple epitopes from multiple antigens to counteract the parasites´ biological complexity. Epitopes were identified by computer-assisted genome-wide screenings, considering sequence conservation criteria, antigens annotation and sub-cellular localization, high binding affinity to antigen presenting molecules, and lack of cross-reactivity to proteins in cattle and other breeding species. We ultimately provide 31 B-cell, 8 CD4 T-cell, and 15 CD8 T-cell epitope sequences from 30 distinct antigens for the prospective design of a genetic ensemble vaccine against the four trypanosome species responsible for African animal trypanosomiasis.
Collapse
|
27
|
Magez S, Pinto Torres JE, Obishakin E, Radwanska M. Infections With Extracellular Trypanosomes Require Control by Efficient Innate Immune Mechanisms and Can Result in the Destruction of the Mammalian Humoral Immune System. Front Immunol 2020; 11:382. [PMID: 32218784 PMCID: PMC7078162 DOI: 10.3389/fimmu.2020.00382] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/18/2020] [Indexed: 12/17/2022] Open
Abstract
Salivarian trypanosomes are extracellular parasites that affect humans, livestock, and game animals around the world. Through co-evolution with the mammalian immune system, trypanosomes have developed defense mechanisms that allow them to thrive in blood, lymphoid vessels, and tissue environments such as the brain, the fat tissue, and testes. Trypanosomes have developed ways to circumvent antibody-mediated killing and block the activation of the lytic arm of the complement pathway. Hence, this makes the innate immune control of the infection a crucial part of the host-parasite interaction, determining infection susceptibility, and parasitemia control. Indeed, trypanosomes use a combination of several independent mechanisms to avoid clearance by the humoral immune system. First, perpetuated antigenic variation of the surface coat allows to escape antibody-mediated elimination. Secondly, when antibodies bind to the coat, they are efficiently transported toward the endocytosis pathway, where they are removed from the coat proteins. Finally, trypanosomes engage in the active destruction of the mammalian humoral immune response. This provides them with a rescue solution in case antigenic variation does not confer total immunological invisibility. Both antigenic variation and B cell destruction pose significant hurdles for the development of anti-trypanosome vaccine strategies. However, developing total immune escape capacity and unlimited growth capabilities within a mammalian host is not beneficial for any parasite, as it will result in the accelerated death of the host itself. Hence, trypanosomes have acquired a system of quorum sensing that results in density-dependent population growth arrest in order to prevent overpopulating the host. The same system could possibly sense the infection-associated host tissue damage resulting from inflammatory innate immune responses, in which case the quorum sensing serves to prevent excessive immunopathology and as such also promotes host survival. In order to put these concepts together, this review summarizes current knowledge on the interaction between trypanosomes and the mammalian innate immune system, the mechanisms involved in population growth regulation, antigenic variation and the immuno-destructive effect of trypanosomes on the humoral immune system. Vaccine trials and a discussion on the role of innate immune modulation in these trials are discussed at the end.
Collapse
Affiliation(s)
- Stefan Magez
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Emmanuel Obishakin
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Biotechnology Division, National Veterinary Research Institute, Vom, Nigeria
| | - Magdalena Radwanska
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
28
|
Establishment of a Standardized Vaccine Protocol for the Analysis of Protective Immune Responses During Experimental Trypanosome Infections in Mice. Methods Mol Biol 2020; 2116:721-738. [PMID: 32221951 DOI: 10.1007/978-1-0716-0294-2_42] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To date, trypanosomosis control in humans and animals is achieved by a combination of parasitological screening and treatment. While this approach has successfully brought down the number of reported T. b. gambiense Human African Trypanosomosis (HAT) cases, the method does not offer a sustainable solution for animal trypanosomosis (AT). The main reasons for this are (i) the worldwide distribution of AT, (ii) the wide range of insect vectors involved in transmission of AT, and (iii) the existence of a wildlife parasite reservoir that can serve as a source for livestock reinfection. Hence, in order to control livestock trypanosomosis the only viable long-term solution is an effective antitrypanosome vaccination strategy. Over the last decades, multiple vaccine approaches have been proposed. Despite repeated reports of promising experimental approaches, none of those made it to a field applicable vaccine format. This failure can in part be attributed to flaws in the experimental design that favor a positive laboratory result. This chapter provides a vaccine protocol that should allow for a proper outcome prediction in experimental anti-AT vaccine approaches.
Collapse
|
29
|
Amisigo CM, Antwi CA, Adjimani JP, Gwira TM. In vitro anti-trypanosomal effects of selected phenolic acids on Trypanosoma brucei. PLoS One 2019; 14:e0216078. [PMID: 31048849 PMCID: PMC6497272 DOI: 10.1371/journal.pone.0216078] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/09/2019] [Indexed: 01/04/2023] Open
Abstract
African trypanosomiasis remains a lethal disease to both humans and livestock. The disease persists due to limited drug availability, toxicity and drug resistance, hence the need for a better understanding of the parasite's biology and provision of alternative forms of therapy. In this study, the in vitro effects of phenolic acids were assessed for their trypanocidal activities against Trypanosoma brucei brucei. The effect of the phenolic acids on Trypanosoma brucei brucei was determined by the alamarBlue assay. The cell cycle effects were determined by flow cytometry and parasite morphological analysis was done by microscopy. Effect on cell proliferation was determined by growth kinetic analysis. Reverse Transcriptase quantitative Polymerase Chain Reaction was used to determine expression of iron dependent enzymes and iron distribution determined by atomic absorption spectroscopy. Gallic acid gave an IC50 of 14.2±1.5 μM. Deferoxamine, gallic acid and diminazene aceturate showed a dose dependent effect on the cell viability and the mitochondrion membrane integrity. Gallic acid, deferoxamine and diminazene aceturate caused loss of kinetoplast in 22%, 26% and 82% of trypanosomes respectively and less than 10% increase in the number of trypanosomes in S phase was observed. Gallic acid caused a 0.6 fold decrease, 50 fold increase and 7 fold increase in the expression levels of the transferrin receptor, ribonucleotide reductase and cyclin 2 genes respectively while treatment with deferoxamine and diminazene aceturate also showed differential expressions of the transferrin receptor, ribonucleotide reductase and cyclin 2 genes. The data suggests that gallic acid possibly exerts its effect on T. brucei via iron chelation leading to structural and morphological changes and arrest of the cell cycle. These together provide information on the cell biology of the parasite under iron starved conditions and provide leads into alternative therapeutic approaches in the treatment of African trypanosomiasis.
Collapse
Affiliation(s)
- Cynthia Mmalebna Amisigo
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Christine Achiaa Antwi
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Jonathan Partt Adjimani
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Theresa Manful Gwira
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| |
Collapse
|
30
|
Volpedo G, Costa L, Ryan N, Halsey G, Satoskar A, Oghumu S. Nanoparticulate drug delivery systems for the treatment of neglected tropical protozoan diseases. J Venom Anim Toxins Incl Trop Dis 2019; 25:e144118. [PMID: 31130996 PMCID: PMC6483407 DOI: 10.1590/1678-9199-jvatitd-1441-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/06/2018] [Indexed: 12/13/2022] Open
Abstract
Neglected Tropical Diseases (NTDs) comprise of a group of seventeen infectious
conditions endemic in many developing countries. Among these diseases are three
of protozoan origin, namely leishmaniasis, Chagas disease, and African
trypanosomiasis, caused by the parasites Leishmania spp.,
Trypanosoma cruzi, and Trypanosoma brucei
respectively. These diseases have their own unique challenges which are
associated with the development of effective prevention and treatment methods.
Collectively, these parasitic diseases cause more deaths worldwide than all
other NTDs combined. Moreover, many current therapies for these diseases are
limited in their efficacy, possessing harmful or potentially fatal side effects
at therapeutic doses. It is therefore imperative that new treatment strategies
for these parasitic diseases are developed. Nanoparticulate drug delivery
systems have emerged as a promising area of research in the therapy and
prevention of NTDs. These delivery systems provide novel mechanisms for targeted
drug delivery within the host, maximizing therapeutic effects while minimizing
systemic side effects. Currently approved drugs may also be repackaged using
these delivery systems, allowing for their potential use in NTDs of protozoan
origin. Current research on these novel delivery systems has provided insight
into possible indications, with evidence demonstrating their improved ability to
specifically target pathogens, penetrate barriers within the host, and reduce
toxicity with lower dose regimens. In this review, we will examine current
research on these delivery systems, focusing on applications in the treatment of
leishmaniasis, Chagas disease, and African trypanosomiasis. Nanoparticulate
systems present a unique therapeutic alternative through the repositioning of
existing medications and directed drug delivery.
Collapse
Affiliation(s)
- Greta Volpedo
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA.,Ohio State University, Department of Microbiology, Columbus, OH, 43210, USA
| | - Lourena Costa
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA.,Universidade Federal de Minas Gerais, Faculdade de Medicina, Departamento de Infectologia e Medicina Tropical, Belo Horizonte, MG, Brasil
| | - Nathan Ryan
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA
| | - Gregory Halsey
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA
| | - Abhay Satoskar
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA.,Ohio State University, Department of Microbiology, Columbus, OH, 43210, USA
| | - Steve Oghumu
- Ohio State University Wexner Medical Center, Department of Pathology, Columbus, OH, 43210, USA
| |
Collapse
|
31
|
Ramirez-Barrios R, Reyna-Bello A, Parra O, Valeris R, Tavares-Marques L, Brizard JP, Demettre E, Seveno M, Martinez-Moreno A, Holzmuller P. Trypanosoma vivax infection in sheep: Different patterns of virulence and pathogenicity associated with differentially expressed proteomes. Vet Parasitol 2019; 276S:100014. [PMID: 32904712 PMCID: PMC7458391 DOI: 10.1016/j.vpoa.2019.100014] [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: 02/08/2019] [Revised: 05/22/2019] [Accepted: 05/27/2019] [Indexed: 11/18/2022]
Abstract
Trypanosoma vivax strains exhibit different virulence and pathogenicity patterns. TvMT1 strain showed low virulence and high pathogenicity. TvLIEM176 strain showed high virulence and moderate pathogenicity. Protein expression varies in high virulence/moderate pathogenicity strain vs low virulence/high pathogenicity strain.
Cattle trypanosomosis caused by Trypanosoma vivax is a widely distributed disease in Africa and Latin America. It causes significant losses in the livestock industry and is characterized by fluctuating parasitemia, anemia, fever, lethargy, and weight loss. In this study we evaluated the virulence (capacity to multiply inside the host and to modulate the host response) and pathogenicity (ability to produce disease and/or mortality) patterns of two T. vivax strains (TvMT1 and TvLIEM176) in experimentally-infected sheep and determined the proteins differentially expressed in the proteomes of these two strains. Hematological and clinical parameters were monitored in experimentally-infected versus non-infected sheep for 60 days. All the infected animals developed discernable parasitemia at 3 days post-infection (dpi), and the first parasitemia peak was observed at 6 dpi. The maximum average value of parasitemia was 1.3 × 107 (95% CI, 7.9 × 105–2 × 108) parasites/ml in TvLIEM176-infected animals, and 2.5 × 106 (95% CI, 1.6 × 105–4 × 107) parasites/ml in TvMT1-infected ones. Anemia and clinical manifestations were more severe in the animals infected by TvMT1 strain than in those infected by TvLIEM176. In the proteomic analysis, a total of 29 proteins were identified, of which 14 exhibited significant differences in their expression levels between strains. Proteins with higher expression in TvLIEM176 were: alpha tubulin, beta tubulin, arginine kinase, glucose-regulated protein 78, paraflagellar protein 3, and T-complex protein 1 subunit theta. Proteins with higher expression in TvMT1 were: chaperonin HSP60, T-complex protein 1 subunit alpha, heat shock protein 70, pyruvate kinase, glycerol kinase, inosine-5'-monophosphate dehydrogenase, 73 kDa paraflagellar rod protein, and vacuolar ATP synthase. There was a difference in the virulence and pathogenicity between the T. vivax strains: TvLIEM176 showed high virulence and moderate pathogenicity, whereas TvMT1 showed low virulence and high pathogenicity. The proteins identified in this study are discussed for their potential involvement in strains’ virulence and pathogenicity, to be further defined as biomarkers of severity in T. vivax infections.
Collapse
|
32
|
Mina JGM, Denny PW. Everybody needs sphingolipids, right! Mining for new drug targets in protozoan sphingolipid biosynthesis. Parasitology 2018; 145:134-147. [PMID: 28637533 PMCID: PMC5964470 DOI: 10.1017/s0031182017001081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/15/2017] [Accepted: 05/18/2017] [Indexed: 12/18/2022]
Abstract
Sphingolipids (SLs) are an integral part of all eukaryotic cellular membranes. In addition, they have indispensable functions as signalling molecules controlling a myriad of cellular events. Disruption of either the de novo synthesis or the degradation pathways has been shown to have detrimental effects. The earlier identification of selective inhibitors of fungal SL biosynthesis promised potent broad-spectrum anti-fungal agents, which later encouraged testing some of those agents against protozoan parasites. In this review we focus on the key enzymes of the SL de novo biosynthetic pathway in protozoan parasites of the Apicomplexa and Kinetoplastidae, outlining the divergence and interconnection between host and pathogen metabolism. The druggability of the SL biosynthesis is considered, alongside recent technology advances that will enable the dissection and analyses of this pathway in the parasitic protozoa. The future impact of these advances for the development of new therapeutics for both globally threatening and neglected infectious diseases is potentially profound.
Collapse
Affiliation(s)
- John G M Mina
- Department of Biosciences,Lower Mountjoy,Stockton Road,Durham DH1 3LE,UK
| | - P W Denny
- Department of Biosciences,Lower Mountjoy,Stockton Road,Durham DH1 3LE,UK
| |
Collapse
|