1
|
CDC50 Orthologues in Plasmodium falciparum Have Distinct Roles in Merozoite Egress and Trophozoite Maturation. mBio 2022; 13:e0163522. [PMID: 35862778 PMCID: PMC9426505 DOI: 10.1128/mbio.01635-22] [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] [Indexed: 11/20/2022] Open
Abstract
In model organisms, type IV ATPases (P4-ATPases) require cell division control protein 50 (CDC50) chaperones for their phospholipid flipping activity. In the malaria parasite Plasmodium falciparum, guanylyl cyclase alpha (GCα) is an integral membrane protein that is essential for release (egress) of merozoites from their host erythrocytes. GCα is unusual in that it contains both a C-terminal cyclase domain and an N-terminal P4-ATPase domain of unknown function. We sought to investigate whether any of the three CDC50 orthologues (termed A, B, and C) encoded by P. falciparum are required for GCα function. Using gene tagging and conditional gene disruption, we demonstrate that CDC50B and CDC50C but not CDC50A are expressed in the clinically important asexual blood stages and that CDC50B is a binding partner of GCα whereas CDC50C is the binding partner of another putative P4-ATPase, phospholipid-transporting ATPase 2 (ATP2). Our findings indicate that CDC50B has no essential role for intraerythrocytic parasite maturation but modulates the rate of parasite egress by interacting with GCα for optimal cGMP synthesis. In contrast, CDC50C is essential for blood stage trophozoite maturation. Additionally, we find that the CDC50C-ATP2 complex may influence parasite endocytosis of host cell hemoglobin and consequently hemozoin formation.
Collapse
|
2
|
van der Watt ME, Reader J, Birkholtz LM. Adapt or Die: Targeting Unique Transmission-Stage Biology for Malaria Elimination. Front Cell Infect Microbiol 2022; 12:901971. [PMID: 35755845 PMCID: PMC9218253 DOI: 10.3389/fcimb.2022.901971] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/06/2022] [Indexed: 12/25/2022] Open
Abstract
Plasmodium parasites have a complex life cycle that includes development in the human host as well as the Anopheles vector. Successful transmission of the parasite between its host and vector therefore requires the parasite to balance its investments in asexual replication and sexual reproduction, varying the frequency of sexual commitment to persist within the human host and generate future opportunities for transmission. The transmission window is extended further by the ability of stage V gametocytes to circulate in peripheral blood for weeks, whereas immature stage I to IV gametocytes sequester in the bone marrow and spleen until final maturation. Due to the low gametocyte numbers in blood circulation and with the ease of targeting such life cycle bottlenecks, transmission represents an efficient target for therapeutic intervention. The biological process of Plasmodium transmission is a multistage, multifaceted process and the past decade has seen a much deeper understanding of the molecular mechanisms and regulators involved. Clearly, specific and divergent processes are used during transmission compared to asexual proliferation, which both poses challenges but also opportunities for discovery of transmission-blocking antimalarials. This review therefore presents an update of our molecular understanding of gametocyte and gamete biology as well as the status of transmission-blocking activities of current antimalarials and lead development compounds. By defining the biological components associated with transmission, considerations for the development of new transmission-blocking drugs to target such untapped but unique biology is suggested as an important, main driver for transmission-blocking drug discovery.
Collapse
Affiliation(s)
- Mariëtte E van der Watt
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | - Janette Reader
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa.,Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Lyn-Marié Birkholtz
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa.,Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
3
|
Djidjou-Demasse R, Ducrot A, Mideo N, Texier G. Understanding dynamics of Plasmodium falciparum gametocytes production: Insights from an age-structured model. J Theor Biol 2022; 539:111056. [PMID: 35150720 DOI: 10.1016/j.jtbi.2022.111056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 11/16/2022]
Abstract
Many models of within-host malaria infection dynamics have been formulated since the pioneering work of Anderson et al. in 1989. Biologically, the goal of these models is to understand what governs the severity of infections, the patterns of infectiousness, and the variation thereof across individual hosts. Mathematically, these models are based on dynamical systems, with standard approaches ranging from K-compartments ordinary differential equations (ODEs) to delay differential equations (DDEs), to capture the relatively constant duration of replication and bursting once a parasite infects a host red blood cell. Using malariatherapy data, which offers fine-scale resolution on the dynamics of infection across a number of individual hosts, we compare the fit and robustness of one of these standard approaches (K-compartments ODE) with a partial differential equations (PDEs) model, which explicitly tracks the "age" of an infected cell. While both models perform quite similarly in terms of goodness-of-fit for suitably chosen K, the K-compartments ODE model particularly overestimates parasite densities early on in infections when the number of repeated compartments is not large enough. Finally, the K-compartments ODE model (for suitably chosen K) and the PDE model highlight a strong qualitative connection between the density of transmissible parasite stages (i.e., gametocytes) and the density of host-damaging (and asexually-replicating) parasite stages. This finding provides a simple tool for predicting which hosts are most infectious to mosquitoes -vectors of Plasmodium parasites- which is a crucial component of global efforts to control and eliminate malaria.
Collapse
Affiliation(s)
| | - Arnaud Ducrot
- Normandie Univ., UNIHAVRE, LMAH, FR-CNRS-3335 ISCN, 76600 Le Havre, France
| | - Nicole Mideo
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Canada
| | - Gaëtan Texier
- Aix Marseille Univ., IRD, AP-HM, SSA, VITROME, IHU Méditerranée Infection, Marseille, France; Centre d'Epidémiologie et de Santé Publique des Armées (CESPA), Marseille, France
| |
Collapse
|
4
|
Jimoh RG, Abisoye OA, Uthman MMB. Ensemble Feed-Forward Neural Network and Support Vector Machine for Prediction of Multiclass Malaria Infection. JOURNAL OF INFORMATION AND COMMUNICATION TECHNOLOGY 2021. [DOI: 10.32890/jict2022.21.1.6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Globally, recent research are focused on developing appropriate and robust algorithms to provide a robust healthcare system that is versatile and accurate. Existing malaria models are plagued with low rate of convergence, overfitting, limited generalization due to restriction to binary cases prediction, and proneness to local minimum errors in finding reliable testing output due to complexity of features in the feature space, which is a black box in nature. This study adopted a stacking method of heterogeneous ensemble learning of ArtificialNeural Network (ANN) and Support Vector Machine (SVM) algorithms to predict multiclass, symptomatic, and climatic malaria infection. ANN produced 48.33 percent accuracy, 60.61 percent sensitivity, and 45.58 percent specificity. SVM with Gaussian kernel function gave better performance results of 85.60 percent accuracy, 84.06 percent sensitivity, and 86.09 percent specificity. Consequently, to improve prediction performance, a stacking method was introduced to ensemble SVM with ANN. The proposed ensemble malaria model was tuned on different thresholds at a threshold value of 0.60, the ensemble model gave an optimum accuracy of 99.86 percent, sensitivity 100 percent, specificity 98.68 percent, and mean square error 0.14. The ensemble model experimental results indicated that stacked multiple classifiers produced better results than a single model. This research demonstrated the efficiency of heterogeneous stacking ensemble model on effects of climatic variations on multiclass malaria infection classification. Furthermore, the model reduced complexity, overfitting, low rate of convergence, and proneness to local minimum error problems of multiclass malaria infection in comparison to previous related models.
Collapse
Affiliation(s)
- Rasheed Gbenga Jimoh
- Department of Computer Science, Faculty of Communication and Information Science, University of Ilorin
| | | | - Muhammed Mubashir Babatunde Uthman
- Department of Epidemiology and Community Health, Faculty of Clinical Sciences, College of Health Sciences, University of Ilorin, Nigeria
| |
Collapse
|
5
|
Haiyambo DH, Aleksenko L, Mumbengegwi D, Bock R, Uusiku P, Malleret B, Rénia L, Quaye IK. Children with Plasmodium vivax infection previously observed in Namibia, were Duffy negative and carried a c.136G > A mutation. BMC Infect Dis 2021; 21:856. [PMID: 34418990 PMCID: PMC8380386 DOI: 10.1186/s12879-021-06573-y] [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: 05/24/2021] [Accepted: 08/13/2021] [Indexed: 11/10/2022] Open
Abstract
Background In a previous study, using a molecular approach, we reported the presence of P. vivax in Namibia. Here, we have extended our investigation to the Duffy antigen genetic profile of individuals of the same cohort with and without Plasmodium infections. Methods Participants with P. vivax (n = 3), P. falciparum (n = 23) mono-infections and co-infections of P. vivax/P. falciparum (n = 4), and P. falciparum/P. ovale (n = 3) were selected from seven regions. Participants with similar age but without any Plasmodium infections (n = 47) were also selected from all the regions. Duffy allelic profile was examined using standard PCR followed by sequencing of amplified products. Sequenced samples were also examined for the presence or absence of G125A mutation in codon 42, exon 2. Results All individuals tested carried the − 67 T > C mutation. However, while all P. vivax infected participants carried the c.G125A mutation, 7/28 P. falciparum infected participants and 9/41 of uninfected participants did not have the c.G125A mutation. The exon 2 region surrounding codon 42, had a c.136G > A mutation that was present in all P. vivax infections. The odds ratio for lack of this mutation with P. vivax infections was (OR 0.015, 95% CI 0.001–0.176; p = 0.001). Conclusion We conclude that P. vivax infections previously reported in Namibia, occurred in Duffy negative participants, carrying the G125A mutation in codon 42. The role of the additional mutation c.136 G > A in exon 2 in P. vivax infections, will require further investigations.
Collapse
Affiliation(s)
- Daniel Hosea Haiyambo
- Department of Biochemistry and Microbiology, University of Namibia School of Medicine, Windhoek, Namibia
| | - Larysa Aleksenko
- Department of Clinical Sciences, University of Lund, Lund, Sweden
| | - Davies Mumbengegwi
- Multidisciplinary Research Center, University of Namibia, Windhoek, Namibia
| | - Ronnie Bock
- Department of Biology, University of Namibia, Windhoek, Namibia
| | - Petrina Uusiku
- Ministry of Health and Social Services Department of Biology, National Vector Borne Disease Control Program, Windhoek, Namibia
| | - Benoit Malleret
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.,Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Laurent Rénia
- A*STAR ID Labs, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Isaac Kweku Quaye
- Faculty of Engineering, Computer and Applied Sciences, Regent University College of Science and Technology, Dansoman, P. O. Box DS 1636, Accra, Ghana.
| |
Collapse
|
6
|
Gnangnon B, Duraisingh MT, Buckee CO. Deconstructing the parasite multiplication rate of Plasmodium falciparum. Trends Parasitol 2021; 37:922-932. [PMID: 34119440 DOI: 10.1016/j.pt.2021.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/22/2023]
Abstract
Epidemiological indicators describing population-level malaria transmission dynamics are widely used to guide policy recommendations. However, the determinants of malaria outcomes within individuals are still poorly understood. This conceptual gap partly reflects the fact that there are few indicators that robustly predict the trajectory of individual infections or clinical outcomes. The parasite multiplication rate (PMR) is a widely used indicator for the Plasmodium intraerythrocytic development cycle (IDC), for example, but its relationship to clinical outcomes is complex. Here, we review its calculation and use in P. falciparum malaria research, as well as the parasite and host factors that impact it. We also provide examples of metrics that can help to link within-host dynamics to malaria clinical outcomes when used alongside the PMR.
Collapse
Affiliation(s)
- Bénédicte Gnangnon
- Center for Communicable Diseases Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Immunology & Infectious Diseases Department, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Manoj T Duraisingh
- Immunology & Infectious Diseases Department, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Caroline O Buckee
- Center for Communicable Diseases Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
7
|
Fernandes P, Briquet S, Patarot D, Loubens M, Hoareau-Coudert B, Silvie O. The dimerisable Cre recombinase allows conditional genome editing in the mosquito stages of Plasmodium berghei. PLoS One 2020; 15:e0236616. [PMID: 33044964 PMCID: PMC7549836 DOI: 10.1371/journal.pone.0236616] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/01/2020] [Indexed: 01/18/2023] Open
Abstract
Asexual blood stages of the malaria parasite are readily amenable to genetic modification via homologous recombination, allowing functional studies of parasite genes that are not essential in this part of the life cycle. However, conventional reverse genetics cannot be applied for the functional analysis of genes that are essential during asexual blood-stage replication. Various strategies have been developed for conditional mutagenesis of Plasmodium, including recombinase-based gene deletion, regulatable promoters, and mRNA or protein destabilization systems. Among these, the dimerisable Cre (DiCre) recombinase system has emerged as a powerful approach for conditional gene deletion in P. falciparum. In this system, the bacteriophage Cre is expressed in the form of two separate, enzymatically inactive polypeptides, each fused to a different rapamycin-binding protein. Rapamycin-induced heterodimerization of the two components restores recombinase activity. We have implemented the DiCre system in the rodent malaria parasite P. berghei, and show that rapamycin-induced excision of floxed DNA sequences can be achieved with very high efficiency in both mammalian and mosquito parasite stages. This tool can be used to investigate the function of essential genes not only in asexual blood stages, but also in other parts of the malaria parasite life cycle.
Collapse
Affiliation(s)
- Priyanka Fernandes
- Centre d’Immunologie et des Maladies Infectieuses, INSERM, CNRS, CIMI-Paris, Sorbonne Université, Paris, France
| | - Sylvie Briquet
- Centre d’Immunologie et des Maladies Infectieuses, INSERM, CNRS, CIMI-Paris, Sorbonne Université, Paris, France
| | - Delphine Patarot
- Centre d’Immunologie et des Maladies Infectieuses, INSERM, CNRS, CIMI-Paris, Sorbonne Université, Paris, France
| | - Manon Loubens
- Centre d’Immunologie et des Maladies Infectieuses, INSERM, CNRS, CIMI-Paris, Sorbonne Université, Paris, France
| | - Bénédicte Hoareau-Coudert
- UMS PASS, Plateforme de Cytométrie de la Pitié-Salpêtrière (CyPS), Sorbonne Université, Paris, France
| | - Olivier Silvie
- Centre d’Immunologie et des Maladies Infectieuses, INSERM, CNRS, CIMI-Paris, Sorbonne Université, Paris, France
- * E-mail:
| |
Collapse
|
8
|
Schmaler M, Orlova-Fink N, Rutishauser T, Abdulla S, Daubenberger C. Human unconventional T cells in Plasmodium falciparum infection. Semin Immunopathol 2020; 42:265-277. [PMID: 32076813 PMCID: PMC7223888 DOI: 10.1007/s00281-020-00791-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 02/07/2020] [Indexed: 12/22/2022]
Abstract
Malaria is an old scourge of humankind and has a large negative impact on the economic development of affected communities. Recent success in malaria control and reduction of mortality seems to have stalled emphasizing that our current intervention tools need to be complemented by malaria vaccines. Different populations of unconventional T cells such as mucosal-associated invariant T (MAIT) cells, invariant natural killer T (iNKT) cells and γδ T cells are gaining attention in the field of malaria immunology. Significant advances in our basic understanding of unconventional T cell biology in rodent malaria models have been made, however, their roles in humans during malaria are less clear. Unconventional T cells are abundant in skin, gut and liver tissues, and long-lasting expansions and functional alterations were observed upon malaria infection in malaria naïve and malaria pre-exposed volunteers. Here, we review the current understanding of involvement of unconventional T cells in anti-Plasmodium falciparum immunity and highlight potential future research avenues.
Collapse
Affiliation(s)
- Mathias Schmaler
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Nina Orlova-Fink
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Tobias Rutishauser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Claudia Daubenberger
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland.
| |
Collapse
|
9
|
Romphosri S, Changruenngam S, Chookajorn T, Modchang C. Role of a Concentration Gradient in Malaria Drug Resistance Evolution: A Combined within- and between-Hosts Modelling Approach. Sci Rep 2020; 10:6219. [PMID: 32277158 PMCID: PMC7148383 DOI: 10.1038/s41598-020-63283-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/26/2020] [Indexed: 12/20/2022] Open
Abstract
Resistance to antimalarial drugs is currently a growing public health problem, resulting in more cases with treatment failure. Although previous studies suggested that a concentration gradient facilitates the antibiotic resistance evolution in bacteria, no attempt has been made to investigate the roles of a concentration gradient in malaria drug resistance. Unlike the person-to-person mode of transmission of bacteria, the malaria parasites need to switch back and forth between the human and mosquito hosts to complete the life cycle and to spread the resistant alleles. Here we developed a stochastic combined within- and between-hosts evolutionary dynamics model specific to malaria parasites in order to investigate the influence of an antimalarial concentration gradient on the evolutionary dynamics of malaria drug resistance. Every stage of malaria development in both human and mosquito hosts are individually modelled using the tau-leaping algorithm. We found that the concentration gradient can accelerate antimalarial resistance evolution. The gain in resistance evolution was improved by the increase in the parasite mutation rate and the mosquito biting rate. In addition, even though the rate of resistance evolution is not sensitive to the changes in parasite reduction ratios (PRRs) of antimalarial drugs, the probability of finding the antimalarial drug resistant parasites decreases when the PRR increases.
Collapse
Affiliation(s)
- Suwat Romphosri
- Biophysics Group, Department of Physics, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Suttikiat Changruenngam
- Biophysics Group, Department of Physics, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Thanat Chookajorn
- Genomics and Evolutionary Medicine Unit (GEM), Center of Excellence in Malaria Research, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Charin Modchang
- Biophysics Group, Department of Physics, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.
- Centre of Excellence in Mathematics, CHE, Bangkok, 10400, Thailand.
- Thailand Center of Excellence in Physics, CHE, 328 Si Ayutthaya Road, Bangkok, 10400, Thailand.
| |
Collapse
|
10
|
Azevedo R, Mendes AM, Prudêncio M. Inhibition of Plasmodium sporogonic stages by ivermectin and other avermectins. Parasit Vectors 2019; 12:549. [PMID: 31752986 PMCID: PMC6873674 DOI: 10.1186/s13071-019-3805-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 11/12/2019] [Indexed: 12/15/2022] Open
Abstract
Background The transmissible forms of Plasmodium parasites result from a process of sporogony that takes place inside their obligatory mosquito vector and culminates in the formation of mammalian-infective parasite forms. Ivermectin is a member of the avermectin family of endectocides, which has been proposed to inhibit malaria transmission due its insecticidal effect. However, it remains unclear whether ivermectin also exerts a direct action on the parasite’s blood and transmission stages. Methods We employed a rodent model of infection to assess the impact of ivermectin treatment on P. berghei asexual and sexual blood forms in vivo. We then made use of a newly established luminescence-based methodology to evaluate the activity of ivermectin and other avermectins against the sporogonic stages of P. berghei parasites in vitro independent of their role on mosquito physiology. Results Our results show that whereas ivermectin does not affect the parasite’s parasitemia, gametocytemia or exflagellation in the mammalian host, several members of the avermectin family of compounds exert a strong inhibitory effect on the generation and development of P. berghei oocysts. Conclusions Our results shed light on the action of avermectins against Plasmodium transmission stages and highlight the potential of these compounds to help prevent the spread of malaria.
Collapse
Affiliation(s)
- Raquel Azevedo
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisbon, Portugal
| | - António M Mendes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisbon, Portugal.
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisbon, Portugal.
| |
Collapse
|
11
|
Host Cytoskeleton Remodeling throughout the Blood Stages of Plasmodium falciparum. Microbiol Mol Biol Rev 2019; 83:83/4/e00013-19. [PMID: 31484690 DOI: 10.1128/mmbr.00013-19] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The asexual intraerythrocytic development of Plasmodium falciparum, causing the most severe form of human malaria, is marked by extensive host cell remodeling. Throughout the processes of invasion, intracellular development, and egress, the erythrocyte membrane skeleton is remodeled by the parasite as required for each specific developmental stage. The remodeling is facilitated by a plethora of exported parasite proteins, and the erythrocyte membrane skeleton is the interface of most of the observed interactions between the parasite and host cell proteins. Host cell remodeling has been extensively described and there is a vast body of information on protein export or the description of parasite-induced structures such as Maurer's clefts or knobs on the host cell surface. Here we specifically review the molecular level of each host cell-remodeling step at each stage of the intraerythrocytic development of P. falciparum We describe key events, such as invasion, knob formation, and egress, and identify the interactions between exported parasite proteins and the host cell cytoskeleton. We discuss each remodeling step with respect to time and specific requirement of the developing parasite to explain host cell remodeling in a stage-specific manner. Thus, we highlight the interaction with the host membrane skeleton as a key event in parasite survival.
Collapse
|
12
|
Khan MD, Thi Vu HH, Lai QT, Ahn JW. Aggravation of Human Diseases and Climate Change Nexus. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E2799. [PMID: 31390751 PMCID: PMC6696070 DOI: 10.3390/ijerph16152799] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 01/24/2023]
Abstract
For decades, researchers have debated whether climate change has an adverse impact on diseases, especially infectious diseases. They have identified a strong relationship between climate variables and vector's growth, mortality rate, reproduction, and spatiotemporal distribution. Epidemiological data further indicates the emergence and re-emergence of infectious diseases post every single extreme weather event. Based on studies conducted mostly between 1990-2018, three aspects that resemble the impact of climate change impact on diseases are: (a) emergence and re-emergence of vector-borne diseases, (b) impact of extreme weather events, and (c) social upliftment with education and adaptation. This review mainly examines and discusses the impact of climate change based on scientific evidences in published literature. Humans are highly vulnerable to diseases and other post-catastrophic effects of extreme events, as evidenced in literature. It is high time that human beings understand the adverse impacts of climate change and take proper and sustainable control measures. There is also the important requirement for allocation of effective technologies, maintenance of healthy lifestyles, and public education.
Collapse
Affiliation(s)
- Mohd Danish Khan
- Resources Recycling Department, University of Science and Technology, (UST), 217, Gajeong-ro, Yuseong-gu, Daejeon-34113, Korea
- Center for Carbon Mineralization, Mineral Resources Research Division, Korea Institute of Geosciences and Mineral Resources (KIGAM), 124 Gwahak-ro, Yuseong-gu, Daejeon-34132, Korea
| | - Hong Ha Thi Vu
- Center for Carbon Mineralization, Mineral Resources Research Division, Korea Institute of Geosciences and Mineral Resources (KIGAM), 124 Gwahak-ro, Yuseong-gu, Daejeon-34132, Korea
| | - Quang Tuan Lai
- Resources Recycling Department, University of Science and Technology, (UST), 217, Gajeong-ro, Yuseong-gu, Daejeon-34113, Korea
- Center for Carbon Mineralization, Mineral Resources Research Division, Korea Institute of Geosciences and Mineral Resources (KIGAM), 124 Gwahak-ro, Yuseong-gu, Daejeon-34132, Korea
| | - Ji Whan Ahn
- Center for Carbon Mineralization, Mineral Resources Research Division, Korea Institute of Geosciences and Mineral Resources (KIGAM), 124 Gwahak-ro, Yuseong-gu, Daejeon-34132, Korea.
| |
Collapse
|
13
|
Khoury DS, Aogo R, Randriafanomezantsoa-Radohery G, McCaw JM, Simpson JA, McCarthy JS, Haque A, Cromer D, Davenport MP. Within-host modeling of blood-stage malaria. Immunol Rev 2019; 285:168-193. [PMID: 30129195 DOI: 10.1111/imr.12697] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Malaria infection continues to be a major health problem worldwide and drug resistance in the major human parasite species, Plasmodium falciparum, is increasing in South East Asia. Control measures including novel drugs and vaccines are in development, and contributions to the rational design and optimal usage of these interventions are urgently needed. Infection involves the complex interaction of parasite dynamics, host immunity, and drug effects. The long life cycle (48 hours in the common human species) and synchronized replication cycle of the parasite population present significant challenges to modeling the dynamics of Plasmodium infection. Coupled with these, variation in immune recognition and drug action at different life cycle stages leads to further complexity. We review the development and progress of "within-host" models of Plasmodium infection, and how these have been applied to understanding and interpreting human infection and animal models of infection.
Collapse
Affiliation(s)
| | - Rosemary Aogo
- Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | | | - James M McCaw
- School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia.,Peter Doherty Institute for Infection and Immunity, The Royal Melbourne Hospital and University of Melbourne, Melbourne, VIC, Australia
| | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Ashraful Haque
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | | | | |
Collapse
|
14
|
Plasmodium falciparum histidine triad protein and calmodulin modulates calcium homeostasis and intracellular proteolysis. Biochem Biophys Res Commun 2018; 503:722-728. [DOI: 10.1016/j.bbrc.2018.06.067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 06/13/2018] [Indexed: 10/28/2022]
|
15
|
Abstract
Malaria is the major cause of mortality and morbidity in tropical countries. The causative agent, Plasmodium sp., has a complex life cycle and is armed with various mechanisms which ensure its continuous transmission. Gametocytes represent the sexual stage of the parasite and are indispensable for the transmission of the parasite from the human host to the mosquito. Despite its vital role in the parasite's success, it is the least understood stage in the parasite's life cycle. The presence of gametocytes in asymptomatic populations and induction of gametocytogenesis by most antimalarial drugs warrants further investigation into its biology. With a renewed focus on malaria elimination and advent of modern technology available to biologists today, the field of gametocyte biology has developed swiftly, providing crucial insights into the molecular mechanisms driving sexual commitment. This review will summarise key current findings in the field of gametocyte biology and address the associated challenges faced in malaria detection, control and elimination.
Collapse
|
16
|
Adeniba OO, Corbin EA, Ewoldt RH, Bashir R. Optomechanical microrheology of single adherent cancer cells. APL Bioeng 2018; 2:016108. [PMID: 31069293 PMCID: PMC6481704 DOI: 10.1063/1.5010721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/18/2018] [Indexed: 01/04/2023] Open
Abstract
There is a close relationship between the mechanical properties of cells and their physiological function. Non-invasive measurements of the physical properties of cells, especially of adherent cells, are challenging to perform. Through a non-contact optical interferometric technique, we measure and combine the phase, amplitude, and frequency of vibrating silicon pedestal micromechanical resonant sensors to quantify the "loss tangent" of individual adherent human colon cancer cells (HT-29). The loss tangent, a dimensionless ratio of viscoelastic energy loss and energy storage - a measure of the viscoelasticity of soft materials, obtained through an optical path length model, was found to be 1.88 ± 0.08 for live cells and 4.32 ± 0.13 for fixed cells, revealing significant changes (p < 0.001) in mechanical properties associated with estimated nanoscale cell membrane fluctuations of 3.86 ± 0.2 nm for live cells and 2.87 ± 0.1 nm for fixed cells. By combining these values with the corresponding two-degree-of-freedom Kelvin-Voigt model, we obtain the elastic stiffness and viscous loss associated with each individual cell rather than estimations from a population. The technique is unique as it decouples the heterogeneity of individual cells in our population and further refines the viscoelastic solution space.
Collapse
Affiliation(s)
| | | | - Randy H Ewoldt
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA
| | | |
Collapse
|
17
|
Tannous S, Ghanem E. A bite to fight: front-line innate immune defenses against malaria parasites. Pathog Glob Health 2018; 112:1-12. [PMID: 29376476 DOI: 10.1080/20477724.2018.1429847] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Malaria infection caused by Plasmodium parasites remains a major health burden worldwide especially in the tropics and subtropics. Plasmodium exhibits a complex life cycle whereby it undergoes a series of developmental stages in the Anopheles mosquito vector and the vertebrate human host. Malaria severity is mainly attributed to the genetic complexity of the parasite which is reflected in the sophisticated mechanisms of invasion and evasion that allow it to overcome the immune responses of both its invertebrate and vertebrate hosts. In this review, we aim to provide an updated, clear and concise summary of the literature focusing on the interactions of the vertebrate innate immune system with Plasmodium parasites, namely sporozoites, merozoites, and trophozoites. The roles of innate immune factors, both humoral and cellular, in anti-Plasmodium defense are described with particular emphasis on the contribution of key innate players including neutrophils, macrophages, and natural killer cells to the clearance of liver and blood stage parasites. A comprehensive understanding of the innate immune responses to malaria parasites remains an important goal that would dramatically help improve the design of original treatment strategies and vaccines, both of which are urgently needed to relieve the burden of malaria especially in endemic countries.
Collapse
Affiliation(s)
- Stephanie Tannous
- a Faculty of Natural and Applied Sciences, Department of Sciences , Notre Dame University , Louaize , Lebanon
| | - Esther Ghanem
- a Faculty of Natural and Applied Sciences, Department of Sciences , Notre Dame University , Louaize , Lebanon
| |
Collapse
|
18
|
Curtidor H, Reyes C, Bermúdez A, Vanegas M, Varela Y, Patarroyo ME. Conserved Binding Regions Provide the Clue for Peptide-Based Vaccine Development: A Chemical Perspective. Molecules 2017; 22:molecules22122199. [PMID: 29231862 PMCID: PMC6149789 DOI: 10.3390/molecules22122199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/17/2022] Open
Abstract
Synthetic peptides have become invaluable biomedical research and medicinal chemistry tools for studying functional roles, i.e., binding or proteolytic activity, naturally-occurring regions’ immunogenicity in proteins and developing therapeutic agents and vaccines. Synthetic peptides can mimic protein sites; their structure and function can be easily modulated by specific amino acid replacement. They have major advantages, i.e., they are cheap, easily-produced and chemically stable, lack infectious and secondary adverse reactions and can induce immune responses via T- and B-cell epitopes. Our group has previously shown that using synthetic peptides and adopting a functional approach has led to identifying Plasmodium falciparumconserved regions binding to host cells. Conserved high activity binding peptides’ (cHABPs) physicochemical, structural and immunological characteristics have been taken into account for properly modifying and converting them into highly immunogenic, protection-inducing peptides (mHABPs) in the experimental Aotus monkey model. This article describes stereo–electron and topochemical characteristics regarding major histocompatibility complex (MHC)-mHABP-T-cell receptor (TCR) complex formation. Some mHABPs in this complex inducing long-lasting, protective immunity have been named immune protection-inducing protein structures (IMPIPS), forming the subunit components in chemically synthesized vaccines. This manuscript summarizes this particular field and adds our recent findings concerning intramolecular interactions (H-bonds or π-interactions) enabling proper IMPIPS structure as well as the peripheral flanking residues (PFR) to stabilize the MHCII-IMPIPS-TCR interaction, aimed at inducing long-lasting, protective immunological memory.
Collapse
Affiliation(s)
- Hernando Curtidor
- Colombian Institute of Immunology Foundation (FIDIC Nonprofit-Making Organisation), Bogotá 111321, Colombia.
- School of Medicine and Health Sciences, University of Rosario, Bogotá 111321, Colombia.
| | - César Reyes
- Colombian Institute of Immunology Foundation (FIDIC Nonprofit-Making Organisation), Bogotá 111321, Colombia.
| | - Adriana Bermúdez
- Colombian Institute of Immunology Foundation (FIDIC Nonprofit-Making Organisation), Bogotá 111321, Colombia.
- School of Medicine and Health Sciences, University of Rosario, Bogotá 111321, Colombia.
| | - Magnolia Vanegas
- Colombian Institute of Immunology Foundation (FIDIC Nonprofit-Making Organisation), Bogotá 111321, Colombia.
- School of Medicine and Health Sciences, University of Rosario, Bogotá 111321, Colombia.
| | - Yahson Varela
- Colombian Institute of Immunology Foundation (FIDIC Nonprofit-Making Organisation), Bogotá 111321, Colombia.
- Faculty of Health Sciences, Applied and Environmental Sciences University (UDCA), Bogotá 111321, Colombia.
| | - Manuel E Patarroyo
- Colombian Institute of Immunology Foundation (FIDIC Nonprofit-Making Organisation), Bogotá 111321, Colombia.
- Faculty of Medicine, National University of Colombia, Bogotá 111321, Colombia.
| |
Collapse
|
19
|
Chowdhury P, Sen S, Kanjilal SD, Sengupta S. Genetic structure of two erythrocyte binding antigens of Plasmodium falciparum reveals a contrasting pattern of selection. INFECTION GENETICS AND EVOLUTION 2017; 57:64-74. [PMID: 29128519 DOI: 10.1016/j.meegid.2017.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/18/2017] [Accepted: 11/07/2017] [Indexed: 11/15/2022]
Abstract
Erythrocyte binding antigens 175 (EBA-175) and 140 (EBA-140) play key roles in erythrocyte invasion by binding to glycophorin A (GPA) and C (GPC) respectively in human malaria. Since antigenic variation in malaria endemic region is a major barrier to development of effective vaccine, we explore the nature and pattern of sequence diversity of these two vaccine candidates in Kolkata, India. Population genetic parameters based on parasite sequences representing region II of Pfeba-175 and Pfeba-140 genes were estimated using DnaSP V.5.10 and MEGA version 6.0. A novel molecular docking approach was implemented to assess the binding affinities of Kolkata Pfeba-175 variants with GPA. P. falciparum Kolkata isolates experienced a recent population expansion as documented by negative Tajima's D, Fu & Li's statistics, unimodal mismatch distribution and star-like median-joining network for both loci. Positive selection seemed to play a major role in shaping the diversity of Pfeba-175 (dN/dS=2.45, and McDonald-Kreitman P-value=0.04) with successive accumulation of Q584K/E, E592A and R664S deriving high frequency haplotypes designated here as F2KH3 and F2KH1. In silico molecular docking demonstrated that polypeptides encoded by F2KH1 and F2KH3 were capable of engaging the parasite ligand into energetically favorable interaction with GPA. Our data demonstrated emergence of Pfeba-175 sequences harboring selectively advantageous nonsynonymous substitutions on Pf3D7 sequence background in the Kolkata parasite isolates. A contrasting pattern of Pf3D7-centric expansion of parasite sequences was noted for Pfeba-140. Together, this study provides a firm genetic and biological support favoring a dominant role of EBA-175 in erythrocyte invasion.
Collapse
Affiliation(s)
- Pramita Chowdhury
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700 019, West Bengal, India
| | - Srikanta Sen
- Mitra Tower, Lake Town, Block-A, Kolkata 700 089, India
| | - Sumana Datta Kanjilal
- Department of Pediatric Medicine, Institute of Post Graduate Medical Education & Research, Kolkata, West Bengal, India
| | - Sanghamitra Sengupta
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700 019, West Bengal, India.
| |
Collapse
|
20
|
Profiling molecular factors associated with pyknosis and developmental arrest induced by an opioid receptor antagonist and dihydroartemisinin in Plasmodium falciparum. PLoS One 2017; 12:e0184874. [PMID: 28934264 PMCID: PMC5608265 DOI: 10.1371/journal.pone.0184874] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 09/03/2017] [Indexed: 12/26/2022] Open
Abstract
Malaria continues to be a devastating disease, largely caused by Plasmodium falciparum infection. We investigated the effects of opioid and cannabinoid receptor antagonists on the growth of intraerythrocytic P. falciparum. The delta opioid receptor antagonist 7-benzylidenenaltrexone (BNTX) and the cannabinoid receptor antagonists rimonaband and SR144528 caused growth arrest of the parasite. Notably BNTX and the established antimalarial drug dihydroartemisinin induced prominent pyknosis in parasite cells after a short period of incubation. We compared genome-wide transcriptome profiles in P. falciparum with different degrees of pyknosis in response to drug treatment, and identified 11 transcripts potentially associated with the evoking of pyknosis, of which three, including glutathione reductase (PfGR), triose phosphate transporter (PfoTPT), and a conserved Plasmodium membrane protein, showed markedly different gene expression levels in accordance with the degree of pyknosis. Furthermore, the use of specific inhibitors confirmed PfGR but not PfoTPT as a possible factor contributing to the development of pyknosis. A reduction in total glutathione levels was also detected in association with increased pyknosis. These results further our understanding of the mechanisms responsible for P. falciparum development and the antimalarial activity of dihydroartemisinin, and provide useful information for the development of novel antimalarial agents.
Collapse
|
21
|
Analytical modeling of the mechanics of early invasion of a merozoite into a human erythrocyte. J Biol Phys 2017; 43:471-479. [PMID: 28914402 DOI: 10.1007/s10867-017-9463-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/10/2017] [Indexed: 10/18/2022] Open
Abstract
In this study, we used a continuum model based on contact mechanics to understand the mechanics of merozoite invasion into human erythrocytes. This model allows us to evaluate the indentation force and work as well as the contact pressure between the merozoite and erythrocyte for an early stage of invasion (γ = 10%). The model predicted an indentation force of 1.3e -11N and an indentation work of 1e -18J. The present analytical model can be considered as a useful tool not only for investigations in mechanobiology and biomechanics but also to explore novel therapeutic targets for malaria and other parasite infections.
Collapse
|
22
|
Painter HJ, Carrasquilla M, Llinás M. Capturing in vivo RNA transcriptional dynamics from the malaria parasite Plasmodium falciparum. Genome Res 2017; 27:1074-1086. [PMID: 28416533 PMCID: PMC5453321 DOI: 10.1101/gr.217356.116] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/22/2017] [Indexed: 12/30/2022]
Abstract
To capture the transcriptional dynamics within proliferating cells, methods to differentiate nascent transcription from preexisting mRNAs are desired. One approach is to label newly synthesized mRNA transcripts in vivo through the incorporation of modified pyrimidines. However, the human malaria parasite, Plasmodium falciparum, is incapable of pyrimidine salvage for mRNA biogenesis. To capture cellular mRNA dynamics during Plasmodium development, we engineered parasites that can salvage pyrimidines through the expression of a single bifunctional yeast fusion gene, cytosine deaminase/uracil phosphoribosyltransferase (FCU). We show that expression of FCU allows for the direct incorporation of thiol-modified pyrimidines into nascent mRNAs. Using developmental stage-specific promoters to express FCU-GFP enables the biosynthetic capture and in-depth analysis of mRNA dynamics from subpopulations of cells undergoing differentiation. We demonstrate the utility of this method by examining the transcriptional dynamics of the sexual gametocyte stage transition, a process that is essential to malaria transmission between hosts. Using the pfs16 gametocyte-specific promoter to express FCU-GFP in 3D7 parasites, we found that sexual stage commitment is governed by transcriptional reprogramming and stabilization of a subset of essential gametocyte transcripts. We also measured mRNA dynamics in F12 gametocyte-deficient parasites and demonstrate that the transcriptional program required for sexual commitment and maturation is initiated but likely aborted due to the absence of the PfAP2-G transcriptional regulator and a lack of gametocyte-specific mRNA stabilization. Biosynthetic labeling of Plasmodium mRNAs is incredibly versatile, can be used to measure transcriptional dynamics at any stage of parasite development, and will allow for future applications to comprehensively measure RNA-protein interactions in the malaria parasite.
Collapse
Affiliation(s)
- Heather J Painter
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Manuela Carrasquilla
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania 16802, USA.,Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| |
Collapse
|
23
|
Asahi H, Kobayashi F, Inoue SI, Niikura M, Yagita K, Tolba MEM. Copper Homeostasis for the Developmental Progression of Intraerythrocytic Malarial Parasite. Curr Top Med Chem 2017; 16:3048-3057. [PMID: 26881705 PMCID: PMC5068492 DOI: 10.2174/1568026616999160215151704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/10/2016] [Accepted: 02/20/2016] [Indexed: 01/22/2023]
Abstract
Malaria is one of the world’s most devastating diseases, particularly in the tropics. In humans, Plasmodium falciparum lives mainly within red blood cells, and malaria pathogenesis depends on the red blood cells being infected with the parasite. Non-esterified fatty acids (NEFAs), including cis-9-octadecenoic acid, and phospholipids have been critical for complete parasite growth in serum-free culture, although the efficacy of NEFAs in sustaining the growth of P. falciparum has varied markedly. Hexadecanoic acid and trans-9-octadecenoic acid have arrested development of the parasite, in association with down-regulation of genes encoding copper-binding proteins. Selective removal of Cu+ ions has blockaded completely the ring–trophozoite–schizont progression of the parasite. The importance of copper homeostasis for the developmental progression of P. falciparum has been confirmed by inhibition of copper-binding proteins that regulate copper physiology and function by associating with copper ions. These data have provided strong evidence for a link between healthy copper homeostasis and successive developmental progression of P. falciparum. Perturbation of copper homeostasis may be, thus, instrumental in drug and vaccine development for the malaria medication. We review the importance of copper homeostasis in the asexual growth of P. falciparum in relation to NEFAs, copper-binding proteins, apoptosis, mitochondria, and gene expression.
Collapse
Affiliation(s)
- Hiroko Asahi
- Division of Tropical Diseases and Parasitology, Department of Infectious Diseases, Kyorin University School of Medicine, Tokyo 181 8611, Japan.
| | | | | | | | | | | |
Collapse
|
24
|
Plasmodium Helical Interspersed Subtelomeric (PHIST) Proteins, at the Center of Host Cell Remodeling. Microbiol Mol Biol Rev 2016; 80:905-27. [PMID: 27582258 DOI: 10.1128/mmbr.00014-16] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During the asexual cycle, Plasmodium falciparum extensively remodels the human erythrocyte to make it a suitable host cell. A large number of exported proteins facilitate this remodeling process, which causes erythrocytes to become more rigid, cytoadherent, and permeable for nutrients and metabolic products. Among the exported proteins, a family of 89 proteins, called the Plasmodium helical interspersed subtelomeric (PHIST) protein family, has been identified. While also found in other Plasmodium species, the PHIST family is greatly expanded in P. falciparum. Although a decade has passed since their first description, to date, most PHIST proteins remain uncharacterized and are of unknown function and localization within the host cell, and there are few data on their interactions with other host or parasite proteins. However, over the past few years, PHIST proteins have been mentioned in the literature at an increasing rate owing to their presence at various localizations within the infected erythrocyte. Expression of PHIST proteins has been implicated in molecular and cellular processes such as the surface display of PfEMP1, gametocytogenesis, changes in cell rigidity, and also cerebral and pregnancy-associated malaria. Thus, we conclude that PHIST proteins are central to host cell remodeling, but despite their obvious importance in pathology, PHIST proteins seem to be understudied. Here we review current knowledge, shed light on the definition of PHIST proteins, and discuss these proteins with respect to their localization and probable function. We take into consideration interaction studies, microarray analyses, or data from blood samples from naturally infected patients to combine all available information on this protein family.
Collapse
|
25
|
Perkins SL, Schaer J. A Modern Menagerie of Mammalian Malaria. Trends Parasitol 2016; 32:772-782. [PMID: 27492115 DOI: 10.1016/j.pt.2016.06.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 12/31/2022]
Abstract
Malaria parasites belong to the diverse apicomplexan order Haemospororida and use a variety of vertebrate and dipteran hosts worldwide. Recently, the utilization of molecular methods has resulted in a burst of newly discovered and rediscovered taxa infecting mammalian hosts, particularly in apes, ungulates, and bats. Additional study of these diverse mammal-infecting taxa is crucial for better understanding the evolutionary history of malaria parasites, especially given that most previous comparative phylogenetic analyses have tended to use both limited taxon sampling and a small set of genetic loci, resulting in weakly supported (and sometimes hotly contested) hypotheses. The ability to generate genomic data from these mammalian parasites, even from subpatent infections, will open up exciting prospects for research on malaria parasites.
Collapse
Affiliation(s)
- Susan L Perkins
- Sackler Institute for Comparative Genomics, American Museum of Natural History, 200 Central Park West, NY, NY 10024, USA.
| | - Juliane Schaer
- Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
26
|
Anand G, Reddy KS, Pandey AK, Mian SY, Singh H, Mittal SA, Amlabu E, Bassat Q, Mayor A, Chauhan VS, Gaur D. A novel Plasmodium falciparum rhoptry associated adhesin mediates erythrocyte invasion through the sialic-acid dependent pathway. Sci Rep 2016; 6:29185. [PMID: 27383149 PMCID: PMC4935899 DOI: 10.1038/srep29185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/10/2016] [Indexed: 01/21/2023] Open
Abstract
Erythrocyte invasion by Plasmodium falciparum merozoites is central to blood-stage infection and malaria pathogenesis. This intricate process is coordinated by multiple parasite adhesins that bind erythrocyte receptors and mediate invasion through several alternate pathways. P. falciparum expresses 2700 genes during the blood-stages, of which the identity and function of many remains unknown. Here, we have identified and characterized a novel P. falciparum rhoptry associated adhesin (PfRA) that mediates erythrocyte invasion through the sialic-acid dependent pathway. PfRA appears to play a significant functional role as it is conserved across different Plasmodium species. It is localized in the rhoptries and further translocated to the merozoite surface. Both native and recombinant PfRA specifically bound erythrocytes in a sialic-acid dependent, chymotrypsin and trypsin resistant manner, which was abrogated by PfRA antibodies confirming a role in erythrocyte invasion. PfRA antibodies inhibited erythrocyte invasion and in combination with antibodies against other parasite ligands produced an additive inhibitory effect, thus validating its important role in erythrocyte invasion. We have thus identified a novel P. falciparum adhesin that binds with a sialic acid containing erythrocyte receptor. Our observations substantiate the strategy to block P. falciparum erythrocyte invasion by simultaneously targeting multiple conserved merozoite antigens involved in alternate invasion pathways.
Collapse
Affiliation(s)
- Gaurav Anand
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - K Sony Reddy
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Alok Kumar Pandey
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Syed Yusuf Mian
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Hina Singh
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Shivani Arora Mittal
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Emmanuel Amlabu
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Quique Bassat
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saude de Manhiça (CISM), Maputo, Mozambique
| | - Alfredo Mayor
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saude de Manhiça (CISM), Maputo, Mozambique
| | - Virander Singh Chauhan
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Deepak Gaur
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
27
|
Ogden NH, Lindsay LR. Effects of Climate and Climate Change on Vectors and Vector-Borne Diseases: Ticks Are Different. Trends Parasitol 2016; 32:646-656. [PMID: 27260548 DOI: 10.1016/j.pt.2016.04.015] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/19/2016] [Accepted: 04/21/2016] [Indexed: 01/05/2023]
Abstract
There has been considerable debate as to whether global risk from vector-borne diseases will be impacted by climate change. This has focussed on important mosquito-borne diseases that are transmitted by the vectors from infected to uninfected humans. However, this debate has mostly ignored the biological diversity of vectors and vector-borne diseases. Here, we review how climate and climate change may impact those most divergent of arthropod disease vector groups: multivoltine insects and hard-bodied (ixodid) ticks. We contrast features of the life cycles and behaviour of these arthropods, and how weather, climate, and climate change may have very different impacts on the spatiotemporal occurrence and abundance of vectors, and the pathogens they transmit.
Collapse
Affiliation(s)
- Nick H Ogden
- National Microbiology Laboratory, Public Health Agency of Canada, 3200 Sicotte, Saint-Hyacinthe, Québec, J2S 7C6, Canada; Groupe de Recherche en Épidémiologie des Zoonoses et Santé Publique, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada; National Microbiology Laboratory, Public Health Agency of Canada, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada.
| | - L Robbin Lindsay
- National Microbiology Laboratory, Public Health Agency of Canada, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| |
Collapse
|
28
|
Alves E, Maluf FV, Bueno VB, Guido RVC, Oliva G, Singh M, Scarpelli P, Costa F, Sartorello R, Catalani LH, Brady D, Tewari R, Garcia CRS. Biliverdin targets enolase and eukaryotic initiation factor 2 (eIF2α) to reduce the growth of intraerythrocytic development of the malaria parasite Plasmodium falciparum. Sci Rep 2016; 6:22093. [PMID: 26915471 PMCID: PMC4768138 DOI: 10.1038/srep22093] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/08/2016] [Indexed: 01/09/2023] Open
Abstract
In mammals, haem degradation to biliverdin (BV) through the action of haem oxygenase (HO) is a critical step in haem metabolism. The malaria parasite converts haem into the chemically inert haemozoin to avoid toxicity. We discovered that the knock-out of HO in P. berghei is lethal; therefore, we investigated the function of biliverdin (BV) and haem in the parasite. Addition of external BV and haem to P. falciparum-infected red blood cell (RBC) cultures delays the progression of parasite development. The search for a BV molecular target within the parasites identified P. falciparum enolase (Pf enolase) as the strongest candidate. Isothermal titration calorimetry using recombinant full-length Plasmodium enolase suggested one binding site for BV. Kinetic assays revealed that BV is a non-competitive inhibitor. We employed molecular modelling studies to predict the new binding site as well as the binding mode of BV to P. falciparum enolase. Furthermore, addition of BV and haem targets the phosphorylation of Plasmodium falciparum eIF2α factor, an eukaryotic initiation factor phosphorylated by eIF2α kinases under stress conditions. We propose that BV targets enolase to reduce parasite glycolysis rates and changes the eIF2α phosphorylation pattern as a molecular mechanism for its action.
Collapse
Affiliation(s)
- Eduardo Alves
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil.,Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brasil
| | - Fernando V Maluf
- Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, Brasil
| | - Vânia B Bueno
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, Brasil
| | - Rafael V C Guido
- Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, Brasil
| | - Glaucius Oliva
- Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, Brasil
| | - Maneesh Singh
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil.,Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brasil
| | - Pedro Scarpelli
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil.,Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brasil
| | - Fahyme Costa
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil.,Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brasil
| | - Robson Sartorello
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil
| | - Luiz H Catalani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, Brasil
| | - Declan Brady
- School of Life Sciences, University of Nottingham, UK
| | - Rita Tewari
- School of Life Sciences, University of Nottingham, UK
| | - Celia R S Garcia
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil
| |
Collapse
|
29
|
New Insights into the Parasitoid Parvilucifera sinerae Life Cycle: The Development and Kinetics of Infection of a Bloom-forming Dinoflagellate Host. Protist 2015; 166:677-99. [DOI: 10.1016/j.protis.2015.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 09/01/2015] [Accepted: 09/12/2015] [Indexed: 11/20/2022]
|
30
|
Josling GA, Llinás M. Sexual development in Plasmodium parasites: knowing when it's time to commit. Nat Rev Microbiol 2015; 13:573-87. [DOI: 10.1038/nrmicro3519] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
31
|
Susi H, Vale PF, Laine AL. Host Genotype and Coinfection Modify the Relationship of within and between Host Transmission. Am Nat 2015; 186:252-63. [PMID: 26655153 DOI: 10.1086/682069] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Variation in individual-level disease transmission is well documented, but the underlying causes of this variation are challenging to disentangle in natural epidemics. In general, within-host replication is critical in determining the extent to which infected hosts shed transmission propagules, but which factors cause variation in this relationship are poorly understood. Here, using a plant host, Plantago lanceolata, and the powdery mildew fungus Podosphaera plantaginis, we quantify how the distinct stages of within-host spread (autoinfection), spore release, and successful transmission to new hosts (alloinfection) are influenced by host genotype, pathogen genotype, and the coinfection status of the host. We find that within-host spread alone fails to predict transmission rates, as this relationship is modified by genetic variation in hosts and pathogens. Their contributions change throughout the course of the epidemic. Host genotype and coinfection had particularly pronounced effects on the dynamics of spore release from infected hosts. Confidently predicting disease spread from local levels of individual transmission, therefore, requires a more nuanced understanding of genotype-specific infection outcomes. This knowledge is key to better understanding the drivers of epidemiological dynamics and the resulting evolutionary trajectories of infectious disease.
Collapse
Affiliation(s)
- Hanna Susi
- Metapopulation Research Group, Department of Biosciences, University of Helsinki, P.O. Box 65 (Viikinkaari 1), FI-00014 Helsinki, Finland
| | | | | |
Collapse
|
32
|
DNA repair mechanisms and their biological roles in the malaria parasite Plasmodium falciparum. Microbiol Mol Biol Rev 2015; 78:469-86. [PMID: 25184562 DOI: 10.1128/mmbr.00059-13] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Research into the complex genetic underpinnings of the malaria parasite Plasmodium falciparum is entering a new era with the arrival of site-specific genome engineering. Previously restricted only to model systems but now expanded to most laboratory organisms, and even to humans for experimental gene therapy studies, this technology allows researchers to rapidly generate previously unattainable genetic modifications. This technological advance is dependent on DNA double-strand break repair (DSBR), specifically homologous recombination in the case of Plasmodium. Our understanding of DSBR in malaria parasites, however, is based largely on assumptions and knowledge taken from other model systems, which do not always hold true in Plasmodium. Here we describe the causes of double-strand breaks, the mechanisms of DSBR, and the differences between model systems and P. falciparum. These mechanisms drive basic parasite functions, such as meiosis, antigen diversification, and copy number variation, and allow the parasite to continually evolve in the contexts of host immune pressure and drug selection. Finally, we discuss the new technologies that leverage DSBR mechanisms to accelerate genetic investigations into this global infectious pathogen.
Collapse
|
33
|
Etiopathogenesis and Pathophysiology of Malaria. HUMAN AND MOSQUITO LYSOZYMES 2015. [PMCID: PMC7123976 DOI: 10.1007/978-3-319-09432-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Malaria is a parasitic disease caused by Plasmodium protozoan parasites and transmitted by Anopheles mosquitoes. The disease is diffused in tropical areas, where it is associated with high morbidity and mortality. P. falciparum is the most dangerous species, mainly affecting young children. The parasite cycle occurs both in humans (asexual stages) and in mosquitoes (sexual stages). In humans, Plasmodium grows and multiplies within red blood cells using hemoglobin as essential source of nutrients and energy. However, this process generates toxic heme that the parasite aggregates into an insoluble inert biocrystal called hemozoin. This molecule sequesters in various organs (liver, spleen, and brain), potentially contributing to the development of malaria immunopathogenesis. Uncomplicated falciparum malaria clinical frame ranges from asymptomatic infection to classic symptoms such as fever, chills, sweating, headache, and muscle aches. However, malaria can also evolve into severe life-threatening complications, including cerebral malaria, severe anemia, respiratory distress, and acute renal failure.
Collapse
|
34
|
Barrera V, Hiscott PS, Craig AG, White VA, Milner DA, Beare NAV, MacCormick IJC, Kamiza S, Taylor TE, Molyneux ME, Harding SP. Severity of retinopathy parallels the degree of parasite sequestration in the eyes and brains of malawian children with fatal cerebral malaria. J Infect Dis 2014; 211:1977-86. [PMID: 25351204 PMCID: PMC4442623 DOI: 10.1093/infdis/jiu592] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/10/2014] [Indexed: 11/17/2022] Open
Abstract
Background. Malarial retinopathy (MR) has diagnostic and prognostic value in children with Plasmodium falciparum cerebral malaria (CM). A clinicopathological correlation between observed retinal changes during life and the degree of sequestration of parasitized red blood cells was investigated in ocular and cerebral vessels at autopsy. Methods. In 18 Malawian children who died from clinically defined CM, we studied the intensity of sequestration and the maturity of sequestered parasites in the retina, in nonretinal ocular tissues, and in the brain. Results. Five children with clinically defined CM during life had other causes of death identified at autopsy, no MR, and scanty intracerebral sequestration. Thirteen children had MR and died from CM. MR severity correlated with percentage of microvessels parasitized in the retina, brain, and nonretinal tissues with some neuroectodermal components (all P < .01). In moderate/severe MR cases (n = 8), vascular congestion was more intense (ρ = 0.841; P < .001), sequestered parasites were more mature, and the quantity of extraerythrocytic hemozoin was higher, compared with mild MR cases (n = 5). Conclusions. These data provide a histopathological basis for the known correlation between degrees of retinopathy and cerebral dysfunction in CM. In addition to being a valuable tool for clinical diagnosis, retinal observations give important information about neurovascular pathophysiology in pediatric CM.
Collapse
Affiliation(s)
- Valentina Barrera
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool
| | - Paul Stephenson Hiscott
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool
| | | | - Valerie Ann White
- Department of Pathology and Laboratory Medicine Department of Ophthalmology and Visual Science, University of British Columbia and Vancouver General Hospital, Canada
| | - Danny Arnold Milner
- Anatomic and Clinical Pathology, Brigham and Women's Hospital Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts
| | - Nicholas Alexander Venton Beare
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool St. Paul's Eye Unit, Royal Liverpool University Hospital, United Kingdom
| | - Ian James Callum MacCormick
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool Malawi-Liverpool-Wellcome Trust Clinical Research Programme
| | | | - Terrie Ellen Taylor
- Department of Osteopathic Medical Specialties, College of Osteopathic Medicine, Michigan State University, East Lansing Blantyre Malaria Project, College of Medicine, University of Malawi, Blantyre
| | - Malcolm Edward Molyneux
- Liverpool School of Tropical Medicine Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts
| | - Simon Peter Harding
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool
| |
Collapse
|
35
|
Njoroge M, Njuguna NM, Mutai P, Ongarora DSB, Smith PW, Chibale K. Recent approaches to chemical discovery and development against malaria and the neglected tropical diseases human African trypanosomiasis and schistosomiasis. Chem Rev 2014; 114:11138-63. [PMID: 25014712 DOI: 10.1021/cr500098f] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | | | - Paul W Smith
- Novartis Institute for Tropical Diseases , Singapore 138670, Singapore
| | | |
Collapse
|
36
|
Frevert U, Nacer A. Immunobiology of Plasmodium in liver and brain. Parasite Immunol 2014; 35:267-82. [PMID: 23631610 DOI: 10.1111/pim.12039] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 04/17/2013] [Indexed: 12/11/2022]
Abstract
Malaria remains one of the most serious health problems globally, but our understanding of the biology of the parasite and the pathogenesis of severe disease is still limited. Multiple cellular effector mechanisms that mediate parasite elimination from the liver have been described, but how effector cells use classical granule-mediated cytotoxicity to attack infected hepatocytes and how cytokines and chemokines spread via the unique fluid pathways of the liver to reach the parasites over considerable distances remains unknown. Similarly, a wealth of information on cerebral malaria (CM), one of the most severe manifestations of the disease, was gained from post-mortem analyses of human brain and murine disease models, but the cellular processes that ultimately cause disease are not fully understood. Here, we discuss how imaging of the local dynamics of parasite infection and host response as well as consideration of anatomical and physiological features of liver and brain can provide a better understanding of the initial asymptomatic hepatic phase of the infection and the cascade of events leading to CM. Given the increasing drug resistance of both parasite and vector and the unavailability of a protective vaccine, the urgency to reduce the tremendous morbidity and mortality associated with severe malaria is obvious.
Collapse
Affiliation(s)
- U Frevert
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine, New York, NY 10010, USA.
| | | |
Collapse
|
37
|
Furuyama W, Enomoto M, Mossaad E, Kawai S, Mikoshiba K, Kawazu SI. An interplay between 2 signaling pathways: melatonin-cAMP and IP3-Ca2+ signaling pathways control intraerythrocytic development of the malaria parasite Plasmodium falciparum. Biochem Biophys Res Commun 2014; 446:125-31. [PMID: 24607908 DOI: 10.1016/j.bbrc.2014.02.070] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/15/2014] [Indexed: 11/26/2022]
Abstract
Plasmodium falciparum spends most of its asexual life cycle within human erythrocytes, where proliferation and maturation occur. Development into the mature forms of P. falciparum causes severe symptoms due to its distinctive sequestration capability. However, the physiological roles and the molecular mechanisms of signaling pathways that govern development are poorly understood. Our previous study showed that P. falciparum exhibits stage-specific spontaneous Calcium (Ca(2+)) oscillations in ring and early trophozoites, and the latter was essential for parasite development. In this study, we show that luzindole (LZ), a selective melatonin receptor antagonist, inhibits parasite growth. Analyses of development and morphology of LZ-treated P. falciparum revealed that LZ severely disrupted intraerythrocytic maturation, resulting in parasite death. When LZ was added at ring stage, the parasite could not undergo further development, whereas LZ added at the trophozoite stage inhibited development from early into late schizonts. Live-cell Ca(2+) imaging showed that LZ treatment completely abolished Ca(2+) oscillation in the ring forms while having little effect on early trophozoites. Further, the melatonin-induced cAMP increase observed at ring and late trophozoite stage was attenuated by LZ treatment. These suggest that a complex interplay between IP3-Ca(2+) and cAMP signaling pathways is involved in intraerythrocytic development of P. falciparum.
Collapse
Affiliation(s)
- Wakako Furuyama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Masahiro Enomoto
- Princess Margaret Cancer Centre, Department of Medical Biophysics, University of Toronto, M5G1L7 Toronto, Ontario, Canada
| | - Ehab Mossaad
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Satoru Kawai
- Laboratory of Tropical Medicine and Parasitology, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Shin-ichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan.
| |
Collapse
|
38
|
RH5-Basigin interaction plays a major role in the host tropism of Plasmodium falciparum. Proc Natl Acad Sci U S A 2013; 110:20735-40. [PMID: 24297912 DOI: 10.1073/pnas.1320771110] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Plasmodium falciparum, the cause of almost all human malaria mortality, is a member of the Laverania subgenus which infects African great apes. Interestingly, Laverania parasites exhibit strict host specificity in their natural environment: P. reichenowi, P. billcollinsi, and P. gaboni infect only chimpanzees; P. praefalciparum, P. blacklocki, and P. adleri are restricted to gorillas, and P. falciparum is pandemic in humans. The molecular mechanism(s) responsible for these host restrictions are not understood, although the interaction between the parasite blood-stage invasion ligand EBA175 and the host erythrocyte receptor Glycophorin-A (GYPA) has been implicated previously. We reexamined the role of the EBA175-GYPA interaction in host tropism using recombinant proteins and biophysical assays and found that EBA175 orthologs from the chimpanzee-restricted parasites P. reichenowi and P. billcollinsi both bound to human GYPA with affinities similar to that of P. falciparum, suggesting that the EBA175-GYPA interaction is unlikely to be the sole determinant of Laverania host specificity. We next investigated the contribution of the recently discovered Reticulocyte-binding protein Homolog 5 (RH5)-Basigin (BSG) interaction in host-species selectivity and found that P. falciparum RH5 bound chimpanzee BSG with a significantly lower affinity than human BSG and did not bind gorilla BSG, mirroring the known host tropism of P. falciparum. Using site-directed mutagenesis, we identified residues in BSG that are responsible for the species specificity of PfRH5 binding. Consistent with the essential role of the PfRH5-BSG interaction in erythrocyte invasion, we conclude that species-specific differences in the BSG receptor provide a molecular explanation for the restriction of P. falciparum to its human host.
Collapse
|
39
|
Shahinas D, Folefoc A, Pillai DR. Targeting Plasmodium falciparum Hsp90: Towards Reversing Antimalarial Resistance. Pathogens 2013; 2:33-54. [PMID: 25436880 PMCID: PMC4235713 DOI: 10.3390/pathogens2010033] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 01/18/2013] [Accepted: 01/23/2013] [Indexed: 12/29/2022] Open
Abstract
Malaria continues to exact a great human toll in tropical settings. Antimalarial resistance is rife and the parasite inexorably develops mechanisms to outwit our best drugs, including the now first-line choice, artesunate. Novel strategies to circumvent resistance are needed. Here we detail drug development focusing on heat shock protein 90 and its central role as a chaperone. A growing body of evidence supports the role for Hsp90 inhibitors as adjunctive drugs able to restore susceptibility to traditionally efficacious compounds like chloroquine.
Collapse
Affiliation(s)
- Dea Shahinas
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Canada.
| | - Asongna Folefoc
- Department of Pathology & Laboratory Medicine, The University of Calgary, Calgary, AB, Canada.
| | - Dylan R Pillai
- Department of Pathology & Laboratory Medicine, The University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
40
|
Hoeijmakers WAM, Salcedo-Amaya AM, Smits AH, Françoijs KJ, Treeck M, Gilberger TW, Stunnenberg HG, Bártfai R. H2A.Z/H2B.Z double-variant nucleosomes inhabit the AT-rich promoter regions of the Plasmodium falciparum genome. Mol Microbiol 2013; 87:1061-73. [PMID: 23320541 PMCID: PMC3594968 DOI: 10.1111/mmi.12151] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2013] [Indexed: 02/06/2023]
Abstract
Histone variants are key components of the epigenetic code and evolved to perform specific functions in transcriptional regulation, DNA repair, chromosome segregation and other fundamental processes. Although variants for histone H2A and H3 are found throughout the eukaryotic kingdom, variants of histone H2B and H4 are rarely encountered. H2B.Z is one of those rare H2B variants and is apicomplexan-specific. Here we show that in Plasmodium falciparum H2B.Z localizes to euchromatic intergenic regions throughout intraerythrocytic development and together with H2A.Z forms a double-variant nucleosome subtype. These nucleosomes are enriched in promoters over 3′ intergenic regions and their occupancy generally correlates with the strength of the promoter, but not with its temporal activity. Remarkably, H2B.Z occupancy levels exhibit a clear correlation with the base-composition of the underlying DNA, raising the intriguing possibility that the extreme AT content of the intergenic regions within the Plasmodium genome might be instructive for histone variant deposition. In summary, our data show that the H2A.Z/H2B.Z double-variant nucleosome demarcates putative regulatory regions of the P. falciparum epigenome and likely provides a scaffold for dynamic regulation of gene expression in this deadly human pathogen.
Collapse
Affiliation(s)
- Wieteke A M Hoeijmakers
- Department of Molecular Biology, Radboud University, Nijmegen Centre for Molecular Life Sciences, Nijmegen 6525GA, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Ansari A, Tuteja R. Genome wide comparative comprehensive analysis of Plasmodium falciparum MCM family with human host. Commun Integr Biol 2013; 5:607-15. [PMID: 23336032 PMCID: PMC3541329 DOI: 10.4161/cib.21759] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mini chromosome maintenance (MCM) proteins 2-7, a subgroup of the large AAA ATPase family are critically required for eukaryotic DNA replication. These proteins are most likely responsible for unwinding DNA at the replication forks. Besides this function, some MCMs are also involved in other chromosome transactions such as transcription, chromatin remodeling and genome stability. All the MCMs contain a conserved region of ~200 amino acids responsible for nucleotide binding. The importance of MCM proteins is evident by the fact that deregulation of the activity of MCM family of proteins appears to be directly linked to human carcinogenesis. This article will focus on members of this important family of proteins from the malaria parasite Plasmodium falciparum and their comparison with the human host.
Collapse
Affiliation(s)
- Abulaish Ansari
- Malaria Group; International Centre for Genetic Engineering and Biotechnology; Aruna Asaf Ali Marg, New Delhi India
| | | |
Collapse
|
42
|
Hopp CS, Flueck C, Solyakov L, Tobin A, Baker DA. Spatiotemporal and functional characterisation of the Plasmodium falciparum cGMP-dependent protein kinase. PLoS One 2012; 7:e48206. [PMID: 23139764 PMCID: PMC3489689 DOI: 10.1371/journal.pone.0048206] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 09/25/2012] [Indexed: 01/18/2023] Open
Abstract
Signalling by 3′–5′-cyclic guanosine monophosphate (cGMP) exists in virtually all eukaryotes. In the apicomplexan parasite Plasmodium, the cGMP-dependent protein kinase (PKG) has previously been reported to play a critical role in four key stages of the life cycle. The Plasmodium falciparum isoform (PfPKG) is essential for the initiation of gametogenesis and for blood stage schizont rupture and work on the orthologue from the rodent malaria parasite P. berghei (PbPKG) has shown additional roles in ookinete differentiation and motility as well as liver stage schizont development. In the present study, PfPKG expression and subcellular location in asexual blood stages was investigated using transgenic epitope-tagged PfPKG-expressing P. falciparum parasites. In Western blotting experiments and immunofluorescence analysis (IFA), maximal PfPKG expression was detected at the late schizont stage. While IFA suggested a cytosolic location, a degree of overlap with markers of the endoplasmic reticulum (ER) was found and subcellular fractionation showed some association with the peripheral membrane fraction. This broad localisation is consistent with the notion that PfPKG, as with the mammalian orthologue, has numerous cellular substrates. This idea is further supported by the global protein phosphorylation pattern of schizonts which was substantially changed following PfPKG inhibition, suggesting a complex role for PfPKG during schizogony.
Collapse
Affiliation(s)
- Christine S. Hopp
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Christian Flueck
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Lev Solyakov
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom
| | - Andrew Tobin
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom
| | - David A. Baker
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- * E-mail:
| |
Collapse
|
43
|
Kumar S, Yokoyama N, Kim JY, Bork-Mimm S, Inoue N, Xuan X, Igarashi I, Sugimoto C. Theileria equi merozoite antigen-2 interacts with actin molecule of equine erythrocyte during their asexual development. Exp Parasitol 2012; 132:508-12. [PMID: 23047133 DOI: 10.1016/j.exppara.2012.09.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 09/20/2012] [Indexed: 11/17/2022]
Abstract
Theileria equi is a tick-transmitted intraerythrocytic protozoan parasite in equids. Equine merozoite antigen (EMA)-1 and EMA-2 of T. equi have been identified as immunodominant proteins co-expressed on the surface of extra-erythrocytic merozoites. Additionally, only the EMA-2 is shed into the cytoplasm of infected erythrocyte or inside the erythrocytic membrane during their early developmental stage. In this study, we initially performed West-Western blot analysis on Triton X-100-insoluble erythrocytic skeleton collected from a healthy horse, using a glutathione S-transferase (GST)-tagged recombinant EMA-1t or EMA-2t of T. equi. The results indicated positive interactions of actin and band 4.1 molecules in the equine erythrocytic skeleton only with the recombinant EMA-2t. Subsequently, we carried out GST pull-down assay using the recombinant antigens (as above) against solubilized lysate of equine erythrocytic skeleton, and confirmed the co-precipitation of actin molecule with EMA-2t, but not with the EMA-1t. The interaction of EMA-2 with host erythrocytic actin indicated its role in the pathobiology of T. equi infection within host erythrocytes.
Collapse
Affiliation(s)
- Sanjay Kumar
- National Research Centre on Equines, Sirsa Road, Hisar 125 001, Haryana, India
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Malaria and human red blood cells. Med Microbiol Immunol 2012; 201:593-8. [PMID: 22965173 DOI: 10.1007/s00430-012-0272-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 08/28/2012] [Indexed: 12/22/2022]
Abstract
Invasion by the malaria parasite, Plasmodium falciparum, brings about extensive changes in the host red cells. These include loss of the normal discoid shape, increased rigidity of the membrane, elevated permeability to a wide variety of ionic and other species and increased adhesiveness, most notably to endothelial surfaces. These effects facilitate survival of the parasite within the host cell and tend to increase the virulence of disease that includes cerebral malaria and anemia. Numerous proteins secreted by the internalized parasite and interacting with red cell membrane proteins are responsible for the changes occurring to the host cell. Anemia, a serious clinical manifestation of malaria, is due to increased destruction of both infected and uninfected red cells due to membrane alterations, as well as ineffective erythropoiesis. There is very good evidence that various red cell disorders including hemoglobinopathies and hereditary ovalocytosis decrease the virulence of disease following parasite infection. A number of mechanism(s) are likely responsible for the protective effect of various red cell abnormalities including decreased invasion, impaired intraerythrocytic development of the parasites and altered interaction between exported parasite proteins and the red cell membrane skeleton.
Collapse
|
45
|
Hoeijmakers WAM, Flueck C, Françoijs KJ, Smits AH, Wetzel J, Volz JC, Cowman AF, Voss T, Stunnenberg HG, Bártfai R. Plasmodium falciparum centromeres display a unique epigenetic makeup and cluster prior to and during schizogony. Cell Microbiol 2012; 14:1391-401. [PMID: 22507744 DOI: 10.1111/j.1462-5822.2012.01803.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 04/05/2012] [Accepted: 04/11/2012] [Indexed: 11/30/2022]
Abstract
Centromeres are essential for the faithful transmission of chromosomes to the next generation, therefore being essential in all eukaryotic organisms. The centromeres of Plasmodium falciparum, the causative agent of the most severe form of malaria, have been broadly mapped on most chromosomes, but their epigenetic composition remained undefined. Here, we reveal that the centromeric histone variant PfCENH3 occupies a 4-4.5 kb region on each P. falciparum chromosome, which is devoid of pericentric heterochromatin but harbours another histone variant, PfH2A.Z. These CENH3 covered regions pinpoint the exact position of the centromere on all chromosomes and revealed that all centromeric regions have similar size and sequence composition. Immunofluorescence assay of PfCENH3 strongly suggests that P. falciparum centromeres cluster to a single nuclear location prior to and during mitosis and cytokinesis but dissociate soon after invasion. In summary, we reveal a dynamic association of Plasmodium centromeres, which bear a unique epigenetic signature and conform to a strict structure. These findings suggest that DNA-associated and epigenetic elements play an important role in centromere establishment in this important human pathogen.
Collapse
Affiliation(s)
- Wieteke A M Hoeijmakers
- Department of Molecular Biology, Radboud University, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hsu YH, Lu P, Coleman JL, Tang WC. A microfluidic platform to isolate avian erythrocytes infected with Plasmodium gallinaceum malaria parasites based on surface morphological changes. Biomed Microdevices 2011; 13:995-1004. [DOI: 10.1007/s10544-011-9569-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
47
|
Abstract
Malaria is caused by intraerythrocytic protozoan parasites belonging to Plasmodium spp. (phylum Apicomplexa) that produce significant morbidity and mortality, mostly in developing countries. Plasmodium parasites have a complex life cycle that includes multiple stages in anopheline mosquito vectors and vertebrate hosts. During the life cycle, the parasites undergo several cycles of extreme population growth within a brief span, and this is critical for their continued transmission and a contributing factor for their pathogenesis in the host. As with other eukaryotes, successful mitosis is an essential requirement for Plasmodium reproduction; however, some aspects of Plasmodium mitosis are quite distinct and not fully understood. In this review, we will discuss the current understanding of the architecture and key events of mitosis in Plasmodium falciparum and related parasites and compare them with the traditional mitotic events described for other eukaryotes.
Collapse
|
48
|
Alves E, Bartlett PJ, Garcia CRS, Thomas AP. Melatonin and IP3-induced Ca2+ release from intracellular stores in the malaria parasite Plasmodium falciparum within infected red blood cells. J Biol Chem 2010; 286:5905-12. [PMID: 21149448 DOI: 10.1074/jbc.m110.188474] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IP(3)-dependent Ca(2+) signaling controls a myriad of cellular processes in higher eukaryotes and similar signaling pathways are evolutionarily conserved in Plasmodium, the intracellular parasite that causes malaria. We have reported that isolated, permeabilized Plasmodium chabaudi, releases Ca(2+) upon addition of exogenous IP(3). In the present study, we investigated whether the IP(3) signaling pathway operates in intact Plasmodium falciparum, the major disease-causing human malaria parasite. P. falciparum-infected red blood cells (RBCs) in the trophozoite stage were simultaneously loaded with the Ca(2+) indicator Fluo-4/AM and caged-IP(3). Photolytic release of IP(3) elicited a transient Ca(2+) increase in the cytosol of the intact parasite within the RBC. The intracellular Ca(2+) pools of the parasite were selectively discharged, using thapsigargin to deplete endoplasmic reticulum (ER) Ca(2+) and the antimalarial chloroquine to deplete Ca(2+) from acidocalcisomes. These data show that the ER is the major IP(3)-sensitive Ca(2+) store. Previous work has shown that the human host hormone melatonin regulates P. falciparum cell cycle via a Ca(2+)-dependent pathway. In the present study, we demonstrate that melatonin increases inositol-polyphosphate production in intact intraerythrocytic parasite. Moreover, the Ca(2+) responses to melatonin and uncaging of IP(3) were mutually exclusive in infected RBCs. Taken together these data provide evidence that melatonin activates PLC to generate IP(3) and open ER-localized IP(3)-sensitive Ca(2+) channels in P. falciparum. This receptor signaling pathway is likely to be involved in the regulation and synchronization of parasite cell cycle progression.
Collapse
Affiliation(s)
- Eduardo Alves
- Department of Pharmacology and Physiology, UMDNJ, New Jersey Medical School, Newark, New Jersey 07103, USA
| | | | | | | |
Collapse
|
49
|
Pollitt LC, Colegrave N, Khan SM, Sajid M, Reece SE. Investigating the evolution of apoptosis in malaria parasites: the importance of ecology. Parasit Vectors 2010; 3:105. [PMID: 21080937 PMCID: PMC3136143 DOI: 10.1186/1756-3305-3-105] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 11/16/2010] [Indexed: 11/16/2022] Open
Abstract
Apoptosis is a precisely regulated process of cell death which occurs widely in multicellular organisms and is essential for normal development and immune defences. In recent years, interest has grown in the occurrence of apoptosis in unicellular organisms. In particular, as apoptosis has been reported in a wide range of species, including protozoan malaria parasites and trypanosomes, it may provide a novel target for intervention. However, it is important to understand when and why parasites employ an apoptosis strategy before the likely long- and short-term success of such an intervention can be evaluated. The occurrence of apoptosis in unicellular parasites provides a challenge for evolutionary theory to explain as organisms are expected to have evolved to maximise their own proliferation, not death. One possible explanation is that protozoan parasites undergo apoptosis in order to gain a group benefit from controlling their density as this prevents premature vector mortality. However, experimental manipulations to examine the ultimate causes behind apoptosis in parasites are lacking. In this review, we focus on malaria parasites to outline how an evolutionary framework can help make predictions about the ecological circumstances under which apoptosis could evolve. We then highlight the ecological considerations that should be taken into account when designing evolutionary experiments involving markers of cell death, and we call for collaboration between researchers in different fields to identify and develop appropriate markers in reference to parasite ecology and to resolve debates on terminology.
Collapse
Affiliation(s)
- Laura C Pollitt
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, School of Biological Sciences, Edinburgh, EH9 3JT, UK
| | - Nick Colegrave
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, School of Biological Sciences, Edinburgh, EH9 3JT, UK
| | - Shahid M Khan
- Leiden Malaria Research group, Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Mohammed Sajid
- Leiden Malaria Research group, Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Sarah E Reece
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, School of Biological Sciences, Edinburgh, EH9 3JT, UK
- Centre for Immunity, Infection and Evolution, University of Edinburgh, School of Biological Sciences, Edinburgh, EH9 3JT, UK
| |
Collapse
|
50
|
Leykauf K, Treeck M, Gilson PR, Nebl T, Braulke T, Cowman AF, Gilberger TW, Crabb BS. Protein kinase a dependent phosphorylation of apical membrane antigen 1 plays an important role in erythrocyte invasion by the malaria parasite. PLoS Pathog 2010; 6:e1000941. [PMID: 20532217 PMCID: PMC2880582 DOI: 10.1371/journal.ppat.1000941] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 05/05/2010] [Indexed: 11/19/2022] Open
Abstract
Apicomplexan parasites are obligate intracellular parasites that infect a variety of hosts, causing significant diseases in livestock and humans. The invasive forms of the parasites invade their host cells by gliding motility, an active process driven by parasite adhesion proteins and molecular motors. A crucial point during host cell invasion is the formation of a ring-shaped area of intimate contact between the parasite and the host known as a tight junction. As the invasive zoite propels itself into the host-cell, the junction moves down the length of the parasite. This process must be tightly regulated and signalling is likely to play a role in this event. One crucial protein for tight-junction formation is the apical membrane antigen 1 (AMA1). Here we have investigated the phosphorylation status of this key player in the invasion process in the human malaria parasite Plasmodium falciparum. We show that the cytoplasmic tail of P. falciparum AMA1 is phosphorylated at serine 610. We provide evidence that the enzyme responsible for serine 610 phosphorylation is the cAMP regulated protein kinase A (PfPKA). Importantly, mutation of AMA1 serine 610 to alanine abrogates phosphorylation of AMA1 in vivo and dramatically impedes invasion. In addition to shedding unexpected new light on AMA1 function, this work represents the first time PKA has been implicated in merozoite invasion.
Collapse
Affiliation(s)
- Kerstin Leykauf
- Macfarlane Burnet Institute for Medical Research & Public Health, Melbourne, Victoria, Australia
| | - Moritz Treeck
- Bernhard Nocht Institute for Tropical Medicine, Department of Molecular Parasitology, Hamburg, Germany
| | - Paul R. Gilson
- Macfarlane Burnet Institute for Medical Research & Public Health, Melbourne, Victoria, Australia
| | - Thomas Nebl
- Walter & Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Thomas Braulke
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alan F. Cowman
- Walter & Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Tim W. Gilberger
- Bernhard Nocht Institute for Tropical Medicine, Department of Molecular Parasitology, Hamburg, Germany
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Brendan S. Crabb
- Macfarlane Burnet Institute for Medical Research & Public Health, Melbourne, Victoria, Australia
- The University of Melbourne, Victoria, Australia
- Monash University, Victoria, Australia
| |
Collapse
|