1
|
Karumuthil T, Eapen A. Risk of Neglected Zoonotic Malaria in Western Ghats: How the Ecosystem Favors Transmission of an Impending Public Health Threat. Am J Trop Med Hyg 2024; 111:934-939. [PMID: 39255806 PMCID: PMC11542525 DOI: 10.4269/ajtmh.24-0311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/16/2024] [Indexed: 09/12/2024] Open
Abstract
The threat of zoonotic malaria remains largely overlooked in India, particularly in regions such as the Western Ghats (WG), a biodiversity hotspot. The WG has diverse species of non-human primates that serve as reservoir hosts for simian malaria parasites. The presence of the Leucosphyrus group of mosquitoes and other vectors of human malaria makes WG vulnerable to the risk of parasite spillover. Natural and anthropogenic factors have contributed to rampant changes in the WG landscape, leading to increased interaction with the sylvatic hosts and vectors. The simian host-human-vector-environment interactions govern the transmission dynamics of zoonotic malaria; however, our knowledge of these interlinkages, encompassing the effect of anthropogenic disruptions in the WG is limited. The impending threat of zoonotic malaria in India could decelerate progress toward malaria elimination, warranting a comprehensive and systematic investigation of disease dynamics in the WG.
Collapse
Affiliation(s)
- Tulasi Karumuthil
- Field Unit, ICMR-National Institute of Malaria Research, Chennai, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Alex Eapen
- Field Unit, ICMR-National Institute of Malaria Research, Chennai, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
2
|
Muh F, Erwina A, Fitriana F, Syahada JH, Cahya AD, Choe S, Jun H, Garjito TA, Siregar JE, Han JH. Plasmodium cynomolgi: What Should We Know? Microorganisms 2024; 12:1607. [PMID: 39203449 PMCID: PMC11356028 DOI: 10.3390/microorganisms12081607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Even though malaria has markedly reduced its global burden, it remains a serious threat to people living in or visiting malaria-endemic areas. The six Plasmodium species (Plasmodium falciparum, Plasmodium vivax, Plasmodium malariae, Plasmodium ovale curtisi, Plasmodium ovale wallikeri and Plasmodium knowlesi) are known to associate with human malaria by the Anopheles mosquito. Highlighting the dynamic nature of malaria transmission, the simian malaria parasite Plasmodium cynomolgi has recently been transferred to humans. The first human natural infection case of P. cynomolgi was confirmed in 2011, and the number of cases is gradually increasing. It is assumed that it was probably misdiagnosed as P. vivax in the past due to its similar morphological features and genome sequences. Comprehensive perspectives that encompass the relationships within the natural environment, including parasites, vectors, humans, and reservoir hosts (macaques), are required to understand this zoonotic malaria and prevent potential unknown risks to human health.
Collapse
Affiliation(s)
- Fauzi Muh
- Department of Epidemiology and Tropical Diseases, Faculty of Public Health, Universitas Diponegoro, Semarang 50275, Indonesia; (F.M.); (A.E.); (F.F.); (J.H.S.)
| | - Ariesta Erwina
- Department of Epidemiology and Tropical Diseases, Faculty of Public Health, Universitas Diponegoro, Semarang 50275, Indonesia; (F.M.); (A.E.); (F.F.); (J.H.S.)
| | - Fadhila Fitriana
- Department of Epidemiology and Tropical Diseases, Faculty of Public Health, Universitas Diponegoro, Semarang 50275, Indonesia; (F.M.); (A.E.); (F.F.); (J.H.S.)
| | - Jadidan Hada Syahada
- Department of Epidemiology and Tropical Diseases, Faculty of Public Health, Universitas Diponegoro, Semarang 50275, Indonesia; (F.M.); (A.E.); (F.F.); (J.H.S.)
| | - Angga Dwi Cahya
- Department of Environmental Health, Faculty of Public Health, Universitas Diponegoro, Semarang 50275, Indonesia;
| | - Seongjun Choe
- Department of Parasitology, School of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea;
| | - Hojong Jun
- Department of Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Triwibowo Ambar Garjito
- Vector-Borne and Zoonotic Research Group, Research Center for Public Health and Nutrition, National Research and Innovation Agency Indonesia, Salatiga 50721, Indonesia;
| | - Josephine Elizabeth Siregar
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Jalan Raya Bogor Km. 46, Cibinong, Bogor 16911, Indonesia;
| | - Jin-Hee Han
- Department of Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea;
| |
Collapse
|
3
|
Gozalo AS, Robinson CK, Holdridge J, Mahecha OFL, Elkins WR. Overview of Plasmodium spp. and Animal Models in Malaria Research. Comp Med 2024; 74:205-230. [PMID: 38902006 PMCID: PMC11373680 DOI: 10.30802/aalas-cm-24-000019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Malaria is a parasitic disease caused by protozoan species of the genus Plasmodium and transmitted by female mosquitos of the genus Anopheles and other Culicidae. Most of the parasites of the genus Plasmodium are highly species specific with more than 200 species described affecting different species of mammals, birds, and reptiles. Plasmodium species strictly affecting humans are P. falciparum, P. vivax, P. ovale, and P. malariae. More recently, P. knowlesi and other nonhuman primate plasmodia were found to naturally infect humans. Currently, malaria occurs mostly in poor tropical and subtropical areas of the world, and in many of these countries it is the leading cause of illness and death. For more than 100 y, animal models, have played a major role in our understanding of malaria biology. Avian Plasmodium species were the first to be used as models to study human malaria. Malaria parasite biology and immunity were first studied using mainly P. gallinaceum and P. relictum. Rodent malarias, particularly P. berghei and P. yoelii, have been used extensively as models to study malaria in mammals. Several species of Plasmodium from nonhuman primates have been used as surrogate models to study human malaria immunology, pathogenesis, candidate vaccines, and treatments. Plasmodium cynomolgi, P. simiovale, and P. fieldi are important models for studying malaria produced by P. vivax and P. ovale, while P. coatneyi is used as a model for study- ing severe malaria. Other nonhuman primate malarias used in research are P. fragile, P. inui, P. knowlesi, P. simium, and P. brasilianum. Very few nonhuman primate species can develop an infection with human malarias. Macaques in general are resistant to infection with P. falciparum, P. vivax, P. malariae, and P. ovale. Only apes and a few species of New World monkeys can support infection with human malarias. Herein we review the most common, and some less common, avian, reptile, and mammal plasmodia species used as models to study human malaria.
Collapse
Affiliation(s)
- Alfonso S Gozalo
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Christen K Robinson
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Julie Holdridge
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Olga Franco L Mahecha
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - William R Elkins
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
4
|
Nikulkova M, Abdrabou W, Carlton JM, Idaghdour Y. Exploiting integrative metabolomics to study host-parasite interactions in Plasmodium infections. Trends Parasitol 2024; 40:313-323. [PMID: 38508901 PMCID: PMC10994734 DOI: 10.1016/j.pt.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/22/2024]
Abstract
Despite years of research, malaria remains a significant global health burden, with poor diagnostic tests and increasing antimalarial drug resistance challenging diagnosis and treatment. While 'single-omics'-based approaches have been instrumental in gaining insight into the biology and pathogenicity of the Plasmodium parasite and its interaction with the human host, a more comprehensive understanding of malaria pathogenesis can be achieved through 'multi-omics' approaches. Integrative methods, which combine metabolomics, lipidomics, transcriptomics, and genomics datasets, offer a holistic systems biology approach to studying malaria. This review highlights recent advances, future directions, and challenges involved in using integrative metabolomics approaches to interrogate the interactions between Plasmodium and the human host, paving the way towards targeted antimalaria therapeutics and control intervention methods.
Collapse
Affiliation(s)
- Maria Nikulkova
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 11101, USA; Johns Hopkins Malaria Research Institute, Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Wael Abdrabou
- Program in Biology, Division of Science and Mathematics, New York University, Abu Dhabi, United Arab Emirates
| | - Jane M Carlton
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 11101, USA; Johns Hopkins Malaria Research Institute, Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | - Youssef Idaghdour
- Program in Biology, Division of Science and Mathematics, New York University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
5
|
DeBarry JD, Nural MV, Pakala SB, Nayak V, Warrenfeltz S, Humphrey J, Lapp SA, Cabrera-Mora M, Brito CFA, Jiang J, Saney CL, Hankus A, Stealey HM, DeBarry MB, Lackman N, Legall N, Lee K, Tang Y, Gupta A, Trippe ED, Bridger RR, Weatherly DB, Peterson MS, Jiang X, Tran V, Uppal K, Fonseca LL, Joyner CJ, Karpuzoglu E, Cordy RJ, Meyer EVS, Wells LL, Ory DS, Lee FEH, Tirouvanziam R, Gutiérrez JB, Ibegbu C, Lamb TJ, Pohl J, Pruett ST, Jones DP, Styczynski MP, Voit EO, Moreno A, Galinski MR, Kissinger JC. MaHPIC malaria systems biology data from Plasmodium cynomolgi sporozoite longitudinal infections in macaques. Sci Data 2022; 9:722. [PMID: 36433985 PMCID: PMC9700667 DOI: 10.1038/s41597-022-01755-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/10/2022] [Indexed: 11/27/2022] Open
Abstract
Plasmodium cynomolgi causes zoonotic malarial infections in Southeast Asia and this parasite species is important as a model for Plasmodium vivax and Plasmodium ovale. Each of these species produces hypnozoites in the liver, which can cause relapsing infections in the blood. Here we present methods and data generated from iterative longitudinal systems biology infection experiments designed and performed by the Malaria Host-Pathogen Interaction Center (MaHPIC) to delve deeper into the biology, pathogenesis, and immune responses of P. cynomolgi in the Macaca mulatta host. Infections were initiated by sporozoite inoculation. Blood and bone marrow samples were collected at defined timepoints for biological and computational experiments and integrative analyses revolving around primary illness, relapse illness, and subsequent disease and immune response patterns. Parasitological, clinical, haematological, immune response, and -omic datasets (transcriptomics, proteomics, metabolomics, and lipidomics) including metadata and computational results have been deposited in public repositories. The scope and depth of these datasets are unprecedented in studies of malaria, and they are projected to be a F.A.I.R., reliable data resource for decades.
Collapse
Affiliation(s)
- Jeremy D DeBarry
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
| | - Mustafa V Nural
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
| | - Suman B Pakala
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Vishal Nayak
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Cancer Data Science Initiatives, Frederick National Laboratory for Cancer Research, Post Office Box B, Frederick, MD, 21702, USA
| | - Susanne Warrenfeltz
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Jay Humphrey
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Stacey A Lapp
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Monica Cabrera-Mora
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Cristiana F A Brito
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Laboratório de Malária, Instituto René Rachou/Fiocruz Minas, Av. Augusto de Lima 1715, Belo Horizonte, MG, 30190 009, Brazil
| | - Jianlin Jiang
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Celia L Saney
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, 30605, USA
| | - Allison Hankus
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Senior Public Health Informaticist, MITRE Corp, Atlanta, GA, 30345, USA
| | - Hannah M Stealey
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Megan B DeBarry
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
| | - Nicolas Lackman
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
| | - Noah Legall
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Interdisciplinary Disease Ecology Across Scales Research Traineeship Program, Institute of Bioinformatics, Center for the Ecology of Infectious Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Kevin Lee
- Center for Integrative Genomics, School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Yan Tang
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Anuj Gupta
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Valted Seq, 704 Quince Orchard Rd, Gaithersburg, MD, 20878, USA
| | - Elizabeth D Trippe
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Federal Drug Administration, Silver Spring, MD, 20993, USA
| | - Robert R Bridger
- Complex Carbohydrate Research Center, Department of Biochemistry, University of Georgia, Athens, GA, 30602, USA
| | - Daniel Brent Weatherly
- Complex Carbohydrate Research Center, Department of Biochemistry, University of Georgia, Athens, GA, 30602, USA
| | - Mariko S Peterson
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Xuntian Jiang
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - ViLinh Tran
- Division of Pulmonary, Allergy, Critical Care, & Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Karan Uppal
- Division of Pulmonary, Allergy, Critical Care, & Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Luis L Fonseca
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, 32603, USA
| | - Chester J Joyner
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Center for Tropical and Emerging Global Disease, University of Georgia, Athens, GA, 30602, USA
- Center for Vaccines and Immunology, Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Ebru Karpuzoglu
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Department of Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Regina J Cordy
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Department of Biology, Wake Forest University, Winston Salem, NC, 27103, USA
| | - Esmeralda V S Meyer
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Institutional Animal Care and Use Committee, Research Compliance and Research Integrity Office, Emory University, Atlanta, GA, 30322, USA
| | - Lance L Wells
- Complex Carbohydrate Research Center, Department of Biochemistry, University of Georgia, Athens, GA, 30602, USA
| | - Daniel S Ory
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Casma Therapeutics, Cambridge, MA, 02139, USA
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, & Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, 30322, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Juan B Gutiérrez
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Department of Mathematics, Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Chris Ibegbu
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Tracey J Lamb
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Jan Pohl
- Biotechnology Core Facility Branch, Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Sarah T Pruett
- Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- University of Tennessee, Knoxville, TN, 37996, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care, & Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Mark P Styczynski
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Eberhard O Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Alberto Moreno
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Mary R Galinski
- Emory Vaccine Center, Yerkes/Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jessica C Kissinger
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA.
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA.
- Department of Genetics, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
6
|
Plasmodium cynomolgi in humans: current knowledge and future directions of an emerging zoonotic malaria parasite. Infection 2022; 51:623-640. [PMID: 36401673 PMCID: PMC9676733 DOI: 10.1007/s15010-022-01952-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022]
Abstract
Plasmodium cynomolgi (Pcy), a simian malaria parasite, is a recent perfect example of emerging zoonotic transfer in human. This review summarizes the current knowledge on the epidemiology of natural Pcy infections in humans, mosquitoes and monkeys, along with its biological, clinical and drug sensitivity patterns. Knowledge gaps and further studies on Pcy in humans are also discussed. This parasite currently seems to be geographically limited in South-East Asia (SEA) with a global prevalence in human ranging from 0 to 1.4%. The Pcy infections were reported in local SEA populations and European travelers, and range from asymptomatic carriage to mild/moderate attacks with no evidence of pathognomonic clinical and laboratory patterns but with Pcy strain-shaped clinical differences. Geographical distribution and competence of suitable mosquito vectors and non-primate hosts, globalization, climate change, and increased intrusion of humans into the habitat of monkeys are key determinants to emergence of Pcy parasites in humans, along with its expansion outside SEA. Sensitization/information campaigns coupled with training and assessment sessions of microscopists and clinicians on Pcy are greatly needed to improve data on the epidemiology and management of human Pcy infection. There is a need for development of sensitive and specific molecular tools for individual diagnosis and epidemiological studies. The development of safe and efficient anti-hypnozoite drugs is the main therapeutic challenge for controlling human relapsing malaria parasites. Experience gained from P. knowlesi malaria, development of integrated measures and strategies—ideally with components related to human, monkeys, mosquito vectors, and environment—could be very helpful to prevent emergence of Pcy malaria in humans through disruption of transmission chain from monkeys to humans and ultimately contain its expansion in SEA and potential outbreaks in a context of malaria elimination.
Collapse
|
7
|
Peterson MS, Joyner CJ, Lapp SA, Brady JA, Wood JS, Cabrera-Mora M, Saney CL, Fonseca LL, Cheng WT, Jiang J, Soderberg SR, Nural MV, Hankus A, Machiah D, Karpuzoglu E, DeBarry JD, Tirouvanziam R, Kissinger JC, Moreno A, Gumber S, Voit EO, Gutierrez JB, Cordy RJ, Galinski MR. Plasmodium knowlesi Cytoadhesion Involves SICA Variant Proteins. Front Cell Infect Microbiol 2022; 12:888496. [PMID: 35811680 PMCID: PMC9260704 DOI: 10.3389/fcimb.2022.888496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium knowlesi poses a health threat throughout Southeast Asian communities and currently causes most cases of malaria in Malaysia. This zoonotic parasite species has been studied in Macaca mulatta (rhesus monkeys) as a model for severe malarial infections, chronicity, and antigenic variation. The phenomenon of Plasmodium antigenic variation was first recognized during rhesus monkey infections. Plasmodium-encoded variant proteins were first discovered in this species and found to be expressed at the surface of infected erythrocytes, and then named the Schizont-Infected Cell Agglutination (SICA) antigens. SICA expression was shown to be spleen dependent, as SICA expression is lost after P. knowlesi is passaged in splenectomized rhesus. Here we present data from longitudinal P. knowlesi infections in rhesus with the most comprehensive analysis to date of clinical parameters and infected red blood cell sequestration in the vasculature of tissues from 22 organs. Based on the histopathological analysis of 22 tissue types from 11 rhesus monkeys, we show a comparative distribution of parasitized erythrocytes and the degree of margination of the infected erythrocytes with the endothelium. Interestingly, there was a significantly higher burden of parasites in the gastrointestinal tissues, and extensive margination of the parasites along the endothelium, which may help explain gastrointestinal symptoms frequently reported by patients with P. knowlesi malarial infections. Moreover, this margination was not observed in splenectomized rhesus that were infected with parasites not expressing the SICA proteins. This work provides data that directly supports the view that a subpopulation of P. knowlesi parasites cytoadheres and sequesters, likely via SICA variant antigens acting as ligands. This process is akin to the cytoadhesive function of the related variant antigen proteins, namely Erythrocyte Membrane Protein-1, expressed by Plasmodium falciparum.
Collapse
Affiliation(s)
- Mariko S. Peterson
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Chester J. Joyner
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Stacey A. Lapp
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Jessica A. Brady
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA, United States
| | - Jennifer S. Wood
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Monica Cabrera-Mora
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Celia L. Saney
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Luis L. Fonseca
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Wayne T. Cheng
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Jianlin Jiang
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Stephanie R. Soderberg
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Mustafa V. Nural
- Institute of Bioinformatics, University of Georgia, Athens, GA, United States
| | - Allison Hankus
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Deepa Machiah
- Division of Pathology, Yerkes National Primate Research Center, Atlanta, GA, United States
| | - Ebru Karpuzoglu
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Jeremy D. DeBarry
- Institute of Bioinformatics, University of Georgia, Athens, GA, United States
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Jessica C. Kissinger
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
- Institute of Bioinformatics, University of Georgia, Athens, GA, United States
- Department of Genetics, University of Georgia, Athens, GA, United States
| | - Alberto Moreno
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Sanjeev Gumber
- Division of Pathology, Yerkes National Primate Research Center, Atlanta, GA, United States
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, GA, United States
| | - Eberhard O. Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Juan B. Gutierrez
- Department of Mathematics, University of Georgia, Athens, GA, United States
| | - Regina Joice Cordy
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Mary R. Galinski
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
8
|
Abstract
"The Primate Malarias" book has been a uniquely important resource for multiple generations of scientists, since its debut in 1971, and remains pertinent to the present day. Indeed, nonhuman primates (NHPs) have been instrumental for major breakthroughs in basic and pre-clinical research on malaria for over 50 years. Research involving NHPs have provided critical insights and data that have been essential for malaria research on many parasite species, drugs, vaccines, pathogenesis, and transmission, leading to improved clinical care and advancing research goals for malaria control, elimination, and eradication. Whilst most malaria scientists over the decades have been studying Plasmodium falciparum, with NHP infections, in clinical studies with humans, or using in vitro culture or rodent model systems, others have been dedicated to advancing research on Plasmodium vivax, as well as on phylogenetically related simian species, including Plasmodium cynomolgi, Plasmodium coatneyi, and Plasmodium knowlesi. In-depth study of these four phylogenetically related species over the years has spawned the design of NHP longitudinal infection strategies for gathering information about ongoing infections, which can be related to human infections. These Plasmodium-NHP infection model systems are reviewed here, with emphasis on modern systems biological approaches to studying longitudinal infections, pathogenesis, immunity, and vaccines. Recent discoveries capitalizing on NHP longitudinal infections include an advanced understanding of chronic infections, relapses, anaemia, and immune memory. With quickly emerging new technological advances, more in-depth research and mechanistic discoveries can be anticipated on these and additional critical topics, including hypnozoite biology, antigenic variation, gametocyte transmission, bone marrow dysfunction, and loss of uninfected RBCs. New strategies and insights published by the Malaria Host-Pathogen Interaction Center (MaHPIC) are recapped here along with a vision that stresses the importance of educating future experts well trained in utilizing NHP infection model systems for the pursuit of innovative, effective interventions against malaria.
Collapse
Affiliation(s)
- Mary R Galinski
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Vaccine Center, Emory University, Atlanta, GA, USA.
- Emory National Primate Research Center (Yerkes National Primate Research Center), Emory University, Atlanta, GA, USA.
| |
Collapse
|
9
|
Tarimo BB, Nyasembe VO, Ngasala B, Basham C, Rutagi IJ, Muller M, Chhetri SB, Rubinstein R, Juliano JJ, Loya M, Dinglasan RR, Lin JT, Mathias DK. Seasonality and transmissibility of Plasmodium ovale in Bagamoyo District, Tanzania. Parasit Vectors 2022; 15:56. [PMID: 35164867 PMCID: PMC8842944 DOI: 10.1186/s13071-022-05181-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/12/2021] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Plasmodium ovale is a neglected malarial parasite that can form latent hypnozoites in the human liver. Over the last decade, molecular surveillance studies of non-falciparum malaria in Africa have highlighted that P. ovale is circulating below the radar, including areas where Plasmodium falciparum is in decline. To eliminate malaria where P. ovale is endemic, a better understanding of its epidemiology, asymptomatic carriage, and transmission biology is needed. METHODS We performed a pilot study on P. ovale transmission as part of an ongoing study of human-to-mosquito transmission of P. falciparum from asymptomatic carriers. To characterize the malaria asymptomatic reservoir, cross-sectional qPCR surveys were conducted in Bagamoyo, Tanzania, over three transmission seasons. Positive individuals were enrolled in transmission studies of P. falciparum using direct skin feeding assays (DFAs) with Anopheles gambiae s.s. (IFAKARA strain) mosquitoes. For a subset of participants who screened positive for P. ovale on the day of DFA, we incubated blood-fed mosquitoes for 14 days to assess sporozoite development. RESULTS Molecular surveillance of asymptomatic individuals revealed a P. ovale prevalence of 11% (300/2718), compared to 29% (780/2718) for P. falciparum. Prevalence for P. ovale was highest at the beginning of the long rainy season (15.5%, 128/826) in contrast to P. falciparum, which peaked later in both the long and short rainy seasons. Considering that these early-season P. ovale infections were low-density mono-infections (127/128), we speculate many were due to hypnozoite-induced relapse. Six of eight P. ovale-infected asymptomatic individuals who underwent DFAs successfully transmitted P. ovale parasites to A. gambiae. CONCLUSIONS Plasmodium ovale is circulating at 4-15% prevalence among asymptomatic individuals in coastal Tanzania, largely invisible to field diagnostics. A different seasonal peak from co-endemic P. falciparum, the capacity to relapse, and efficient transmission to Anopheles vectors likely contribute to its persistence amid control efforts focused on P. falciparum.
Collapse
Affiliation(s)
- Brian B. Tarimo
- Vector Immunity and Transmission Biology Unit, Department of Environmental Health and Ecological Science, Ifakara Health Institute-Bagamoyo Office, P.O. Box 74, Bagamoyo, Coast Region 61301 Tanzania
| | - Vincent O. Nyasembe
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL 32611 USA
| | - Billy Ngasala
- Department of Parasitology and Medical Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, Dar es Salaam, 11103 Tanzania
| | - Christopher Basham
- Institute of Global Health and Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, NC 27599 USA
| | - Isaack J. Rutagi
- Vector Immunity and Transmission Biology Unit, Department of Environmental Health and Ecological Science, Ifakara Health Institute-Bagamoyo Office, P.O. Box 74, Bagamoyo, Coast Region 61301 Tanzania
| | - Meredith Muller
- Institute of Global Health and Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, NC 27599 USA
| | - Srijana B. Chhetri
- Institute of Global Health and Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, NC 27599 USA
| | - Rebecca Rubinstein
- Institute of Global Health and Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, NC 27599 USA
| | - Jonathan J. Juliano
- Institute of Global Health and Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, NC 27599 USA
| | - Mwajabu Loya
- Department of Parasitology and Medical Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, Dar es Salaam, 11103 Tanzania
| | - Rhoel R. Dinglasan
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL 32611 USA
| | - Jessica T. Lin
- Institute of Global Health and Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, NC 27599 USA
| | - Derrick K. Mathias
- Department of Entomology and Nematology, Florida Medical Entomology Laboratory, Institute of Food and Agricultural Sciences, University of Florida, Vero Beach, FL 32962 USA
| |
Collapse
|
10
|
Peterson MS, Joyner CJ, Brady JA, Wood JS, Cabrera-Mora M, Saney CL, Fonseca LL, Cheng WT, Jiang J, Lapp SA, Soderberg SR, Nural MV, Humphrey JC, Hankus A, Machiah D, Karpuzoglu E, DeBarry JD, Tirouvanziam R, Kissinger JC, Moreno A, Gumber S, Voit EO, Gutiérrez JB, Cordy RJ, Galinski MR. Clinical recovery of Macaca fascicularis infected with Plasmodium knowlesi. Malar J 2021; 20:486. [PMID: 34969401 PMCID: PMC8719393 DOI: 10.1186/s12936-021-03925-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/24/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Kra monkeys (Macaca fascicularis), a natural host of Plasmodium knowlesi, control parasitaemia caused by this parasite species and escape death without treatment. Knowledge of the disease progression and resilience in kra monkeys will aid the effective use of this species to study mechanisms of resilience to malaria. This longitudinal study aimed to define clinical, physiological and pathological changes in kra monkeys infected with P. knowlesi, which could explain their resilient phenotype. METHODS Kra monkeys (n = 15, male, young adults) were infected intravenously with cryopreserved P. knowlesi sporozoites and the resulting parasitaemias were monitored daily. Complete blood counts, reticulocyte counts, blood chemistry and physiological telemetry data (n = 7) were acquired as described prior to infection to establish baseline values and then daily after inoculation for up to 50 days. Bone marrow aspirates, plasma samples, and 22 tissue samples were collected at specific time points to evaluate longitudinal clinical, physiological and pathological effects of P. knowlesi infections during acute and chronic infections. RESULTS As expected, the kra monkeys controlled acute infections and remained with low-level, persistent parasitaemias without anti-malarial intervention. Unexpectedly, early in the infection, fevers developed, which ultimately returned to baseline, as well as mild to moderate thrombocytopenia, and moderate to severe anaemia. Mathematical modelling and the reticulocyte production index indicated that the anaemia was largely due to the removal of uninfected erythrocytes and not impaired production of erythrocytes. Mild tissue damage was observed, and tissue parasite load was associated with tissue damage even though parasite accumulation in the tissues was generally low. CONCLUSIONS Kra monkeys experimentally infected with P. knowlesi sporozoites presented with multiple clinical signs of malaria that varied in severity among individuals. Overall, the animals shared common mechanisms of resilience characterized by controlling parasitaemia 3-5 days after patency, and controlling fever, coupled with physiological and bone marrow responses to compensate for anaemia. Together, these responses likely minimized tissue damage while supporting the establishment of chronic infections, which may be important for transmission in natural endemic settings. These results provide new foundational insights into malaria pathogenesis and resilience in kra monkeys, which may improve understanding of human infections.
Collapse
Affiliation(s)
- Mariko S Peterson
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - Chester J Joyner
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Center for Vaccines and Immunology, Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Center for Vaccines and Immunology, Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Jessica A Brady
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Jennifer S Wood
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Monica Cabrera-Mora
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Celia L Saney
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Luis L Fonseca
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Wayne T Cheng
- Center for Vaccines and Immunology, Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - Jianlin Jiang
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Stacey A Lapp
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Stephanie R Soderberg
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Thermo Fisher Scientific, South San Francisco, CA, USA
| | - Mustafa V Nural
- Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| | - Jay C Humphrey
- Institute of Bioinformatics, University of Georgia, Athens, GA, USA
- Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Allison Hankus
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- The MITRE Corporation, Atlanta, GA, USA
| | - Deepa Machiah
- Division of Pathology, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Ebru Karpuzoglu
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Department of Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Jeremy D DeBarry
- Institute of Bioinformatics, University of Georgia, Athens, GA, USA
- Center for Topical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | | | - Jessica C Kissinger
- Institute of Bioinformatics, University of Georgia, Athens, GA, USA
- Department of Genetics, University of Georgia, Athens, GA, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Alberto Moreno
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Sanjeev Gumber
- Division of Pathology, Yerkes National Primate Research Center, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, GA, USA
- Pathology, Drug Safety, and DMPK, Boehringer Ingelheim Animal Health USA, Inc., Athens, GA, USA
| | - Eberhard O Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Juan B Gutiérrez
- Department of Mathematics, University of Georgia, Athens, GA, USA
- Department of Mathematics, University of Texas at San Antonio, San Antonio, TX, USA
| | - Regina Joice Cordy
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Department of Biology, Wake Forest University, Winston-Salem, NC, USA
| | - Mary R Galinski
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
- Emory Vaccine Center, Emory University, Atlanta, GA, USA.
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
11
|
Vythilingam I, Chua TH, Liew JWK, Manin BO, Ferguson HM. The vectors of Plasmodium knowlesi and other simian malarias Southeast Asia: challenges in malaria elimination. ADVANCES IN PARASITOLOGY 2021; 113:131-189. [PMID: 34620382 DOI: 10.1016/bs.apar.2021.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Plasmodium knowlesi, a simian malaria parasite of great public health concern has been reported from most countries in Southeast Asia and exported to various countries around the world. Currently P. knowlesi is the predominant species infecting humans in Malaysia. Besides this species, other simian malaria parasites such as P. cynomolgi and P. inui are also infecting humans in the region. The vectors of P. knowlesi and other Asian simian malarias belong to the Leucosphyrus Group of Anopheles mosquitoes which are generally forest dwelling species. Continual deforestation has resulted in these species moving into forest fringes, farms, plantations and human settlements along with their macaque hosts. Limited studies have shown that mosquito vectors are attracted to both humans and macaque hosts, preferring to bite outdoors and in the early part of the night. We here review the current status of simian malaria vectors and their parasites, knowledge of vector competence from experimental infections and discuss possible vector control measures. The challenges encountered in simian malaria elimination are also discussed. We highlight key knowledge gaps on vector distribution and ecology that may impede effective control strategies.
Collapse
Affiliation(s)
- Indra Vythilingam
- Department of Parasitology, University of Malaya, Kuala Lumpur, Malaysia.
| | - Tock Hing Chua
- Department of Pathobiology and Microbiology, Faculty of Medicine and Health Sciences, Universiti Sabah Malaysia, Kota Kinabalu, Sabah, Malaysia.
| | - Jonathan Wee Kent Liew
- Department of Parasitology, University of Malaya, Kuala Lumpur, Malaysia; Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Benny O Manin
- Department of Pathobiology and Microbiology, Faculty of Medicine and Health Sciences, Universiti Sabah Malaysia, Kota Kinabalu, Sabah, Malaysia
| | - Heather M Ferguson
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, Scotland, United Kingdom
| |
Collapse
|
12
|
Gupta A, Styczynski MP, Galinski MR, Voit EO, Fonseca LL. Dramatic transcriptomic differences in Macaca mulatta and Macaca fascicularis with Plasmodium knowlesi infections. Sci Rep 2021; 11:19519. [PMID: 34593836 PMCID: PMC8484567 DOI: 10.1038/s41598-021-98024-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/30/2021] [Indexed: 12/02/2022] Open
Abstract
Plasmodium knowlesi, a model malaria parasite, is responsible for a significant portion of zoonotic malaria cases in Southeast Asia and must be controlled to avoid disease severity and fatalities. However, little is known about the host-parasite interactions and molecular mechanisms in play during the course of P. knowlesi malaria infections, which also may be relevant across Plasmodium species. Here we contrast P. knowlesi sporozoite-initiated infections in Macaca mulatta and Macaca fascicularis using whole blood RNA-sequencing and transcriptomic analysis. These macaque hosts are evolutionarily close, yet malaria-naïve M. mulatta will succumb to blood-stage infection without treatment, whereas malaria-naïve M. fascicularis controls parasitemia without treatment. This comparative analysis reveals transcriptomic differences as early as the liver phase of infection, in the form of signaling pathways that are activated in M. fascicularis, but not M. mulatta. Additionally, while most immune responses are initially similar during the acute stage of the blood infection, significant differences arise subsequently. The observed differences point to prolonged inflammation and anti-inflammatory effects of IL10 in M. mulatta, while M. fascicularis undergoes a transcriptional makeover towards cell proliferation, consistent with its recovery. Together, these findings suggest that timely detection of P. knowlesi in M. fascicularis, coupled with control of inflammation while initiating the replenishment of key cell populations, helps contain the infection. Overall, this study points to specific genes and pathways that could be investigated as a basis for new drug targets that support recovery from acute malaria.
Collapse
Affiliation(s)
- Anuj Gupta
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Mark P Styczynski
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Mary R Galinski
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Eberhard O Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| | - Luis L Fonseca
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Laboratory for Systems Medicine, Department of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
13
|
Hang JW, Tukijan F, Lee EQH, Abdeen SR, Aniweh Y, Malleret B. Zoonotic Malaria: Non- Laverania Plasmodium Biology and Invasion Mechanisms. Pathogens 2021; 10:889. [PMID: 34358039 PMCID: PMC8308728 DOI: 10.3390/pathogens10070889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 12/27/2022] Open
Abstract
Malaria, which is caused by Plasmodium parasites through Anopheles mosquito transmission, remains one of the most life-threatening diseases affecting hundreds of millions of people worldwide every year. Plasmodium vivax, which accounts for the majority of cases of recurring malaria caused by the Plasmodium (non-Laverania) subgenus, is an ancient and continuing zoonosis originating from monkey hosts probably outside Africa. The emergence of other zoonotic malarias (P. knowlesi, P. cynomolgi, and P. simium) further highlights the seriousness of the disease. The severity of this epidemic disease is dependent on many factors, including the parasite characteristics, host-parasite interactions, and the pathology of the infection. Successful infection depends on the ability of the parasite to invade the host; however, little is known about the parasite invasion biology and mechanisms. The lack of this information adds to the challenges to malaria control and elimination, hence enhancing the potential for continuation of this zoonosis. Here, we review the literature describing the characteristics, distribution, and genome details of the parasites, as well as host specificity, host-parasite interactions, and parasite pathology. This information will provide the basis of a greater understanding of the epidemiology and pathogenesis of malaria to support future development of strategies for the control and prevention of this zoonotic infection.
Collapse
Affiliation(s)
- Jing-Wen Hang
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore; (J.W.H.); (F.T.); (E.Q.H.L.)
| | - Farhana Tukijan
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore; (J.W.H.); (F.T.); (E.Q.H.L.)
| | - Erica-Qian-Hui Lee
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore; (J.W.H.); (F.T.); (E.Q.H.L.)
| | - Shifana Raja Abdeen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138648, Singapore;
| | - Yaw Aniweh
- West Africa Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Legon, Accra, Ghana;
| | - Benoit Malleret
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore; (J.W.H.); (F.T.); (E.Q.H.L.)
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138648, Singapore;
| |
Collapse
|
14
|
der Wel AVV, Hofman SO, Kocken CHM. Isolation of GFP-expressing Malarial Hypnozoites by Flow Cytometry Cell Sorting. Bio Protoc 2021; 11:e4006. [PMID: 34124306 DOI: 10.21769/bioprotoc.4006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/14/2021] [Accepted: 03/19/2021] [Indexed: 11/02/2022] Open
Abstract
Hypnozoites are dormant liver-stage parasites unique to relapsing malarial species, including the important human pathogen Plasmodium vivax, and pose a barrier to the elimination of malaria. Little is known regarding the biology of these stages, largely due to their inaccessible location. Hypnozoites can be cultured in vitro but these cultures always consist of a mixture of hepatocytes, developing forms, and hypnozoites. Here, using a GFP-expressing line of the hypnozoite model parasite Plasmodium cynomolgi, we describe a protocol for the FACS-based isolation of malarial hypnozoites. The purified hypnozoites can be used for a range of '-omics' studies to dissect the biology of this cryptic stage of the malarial life cycle.
Collapse
Affiliation(s)
| | - Sam O Hofman
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| |
Collapse
|
15
|
Mukherjee P, Burgio G, Heitlinger E. Dual RNA Sequencing Meta-analysis in Plasmodium Infection Identifies Host-Parasite Interactions. mSystems 2021; 6:e00182-21. [PMID: 33879496 PMCID: PMC8546971 DOI: 10.1128/msystems.00182-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/04/2021] [Indexed: 11/20/2022] Open
Abstract
Dual RNA sequencing (RNA-Seq) is the simultaneous transcriptomic analysis of interacting symbionts, for example, in malaria. Potential cross-species interactions identified by correlated gene expression might highlight interlinked signaling, metabolic, or gene regulatory pathways in addition to physically interacting proteins. Often, malaria studies address one of the interacting organisms-host or parasite-rendering the other "contamination." Here we perform a meta-analysis using such studies for cross-species expression analysis. We screened experiments for gene expression from host and Plasmodium. Out of 171 studies in Homo sapiens, Macaca mulatta, and Mus musculus, we identified 63 potential studies containing host and parasite data. While 16 studies (1,950 samples) explicitly performed dual RNA-Seq, 47 (1,398 samples) originally focused on one organism. We found 915 experimental replicates from 20 blood studies to be suitable for coexpression analysis and used orthologs for meta-analysis across different host-parasite systems. Centrality metrics from the derived gene expression networks correlated with gene essentiality in the parasites. We found indications of host immune response to elements of the Plasmodium protein degradation system, an antimalarial drug target. We identified well-studied immune responses in the host with our coexpression networks, as our approach recovers known broad processes interlinked between hosts and parasites in addition to individual host and parasite protein associations. The set of core interactions represents commonalities between human malaria and its model systems for prioritization in laboratory experiments. Our approach might also allow insights into the transferability of model systems for different pathways in malaria studies.IMPORTANCE Malaria still causes about 400,000 deaths a year and is one of the most studied infectious diseases. The disease is studied in mice and monkeys as lab models to derive potential therapeutic intervention in human malaria. Interactions between Plasmodium spp. and its hosts are either conserved across different host-parasite systems or idiosyncratic to those systems. Here we use correlation of gene expression from different RNA-Seq studies to infer common host-parasite interactions across human, mouse, and monkey studies. First, we find a set of very conserved interactors, worth further scrutiny in focused laboratory experiments. Second, this work might help assess to which extent experiments and knowledge on different pathways can be transferred from models to humans for potential therapy.
Collapse
Affiliation(s)
- Parnika Mukherjee
- Department of Molecular Parasitology, Humboldt University, Berlin, Germany
- Research Group Ecology and Evolution of Molecular Parasite-Host Interactions, Leibniz-Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
- Department of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Gaétan Burgio
- Department of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Emanuel Heitlinger
- Department of Molecular Parasitology, Humboldt University, Berlin, Germany
- Research Group Ecology and Evolution of Molecular Parasite-Host Interactions, Leibniz-Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
| |
Collapse
|
16
|
França ACB, Françoso KS, Marques RF, Trossini GHG, Gomes RA, Póvoa MM, Cunha MG, Silveira ELV, Soares IS. Antibodies Against the Plasmodium vivax Apical Membrane Antigen 1 From the Belem Strain Share Common Epitopes Among Other Worldwide Variants. Front Cell Infect Microbiol 2021; 11:616230. [PMID: 33796476 PMCID: PMC8009186 DOI: 10.3389/fcimb.2021.616230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/23/2021] [Indexed: 12/31/2022] Open
Abstract
Malaria is a human parasitic disease distributed in many tropical countries and caused by various Plasmodium species. Plasmodium vivax has the largest geographical distribution of the Plasmodium species and is predominant in the Americas, including Brazil. Only a small number of P. vivax vaccine formulations have successfully reached clinical trials relative to their P. falciparum counterparts. One of the candidate antigens for a blood-stage P. vivax vaccine is apical membrane antigen 1 (PvAMA-1). Due to the worldwide distribution of Plasmodium parasites, a high degree of variability has been detected in this antigen sequence, representing a considerable challenge to the development of a universal vaccine against malaria. In this study, we evaluated how PvAMA-1 polymorphisms influence vaccine-derived immune responses in P. vivax malaria. To this end, we expressed 9 recombinant protein representatives of different PvAMA-1 allelic variants in the yeast Pichia pastoris: Belem, Chesson I, Sal-1, Indonesia XIX, SK0814, TC103, PNG_05_ESP, PNG_62_MU, and PNG_68_MAS. After protein expression and purification, we evaluated the breadth of the immune responses derived from malaria-exposed individuals from the Amazon region. From 611 serum samples of malaria-exposed individuals, 53.68% of them reacted against the PvAMA-1 Belem through ELISA. Positive samples were further tested against recombinant proteins representing the other PvAMA-1 allelic variants. Whereas Sal-1, Chesson I and SK0814 variants were highly recognized by tested serum samples, Indonesia XIX, TC103, PNG_05_ESP, PNG_62_MU, and PNG_68_MAS were only slightly recognized. Moreover, polyclonal sera derived from C57BL/6 mice immunized with the PvAMA-1 Belem protein predominantly recognized Belem, Sal-1, Chesson I, SK0814, and Indonesia XIX through ELISA. Last, ELISA-based competition assays demonstrated that a previous interaction between anti-Belem polyclonal serum and Sal-1, Chesson I, SK0814, or Indonesia XIX proteins could further inhibit antibody binding to the Belem variant. Our human and mouse data suggest the presence of common epitopes or cross-reactivity between Belem, Sal-1, Chesson I, and SK0814 variants. Although the PvAMA-1 Belem variant induces strain-transcendent antibodies, PvAMA-1 variants from Thailand and Papua New Guinea may need to be included in a universal vaccine formulation to achieve protection against P. vivax malaria.
Collapse
Affiliation(s)
- Ana Caroline Barbosa França
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Kátia Sanches Françoso
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rodolfo Ferreira Marques
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gustavo H. G. Trossini
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Renan A. Gomes
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Maristela G. Cunha
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Eduardo L. V. Silveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Irene S. Soares
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Pasini EM, Kocken CHM. Parasite-Host Interaction and Pathophysiology Studies of the Human Relapsing Malarias Plasmodium vivax and Plasmodium ovale Infections in Non-Human Primates. Front Cell Infect Microbiol 2021; 10:614122. [PMID: 33680982 PMCID: PMC7925837 DOI: 10.3389/fcimb.2020.614122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/24/2020] [Indexed: 12/26/2022] Open
Abstract
Malaria remains a serious health concern across the globe. Historically neglected, non-Falciparum human malarias were put back on the agenda by a paradigm shift in the fight against malaria from malaria control to malaria eradication. Here, we review the modeling of the relapsing parasites Plasmodium vivax (P. vivax) and Plasmodium ovale (P. ovale) in non-human primates with a specific focus on the contribution of these models to our current understanding of the factors that govern parasite-host interactions in P. vivax and P. ovale parasite biology and pathophysiology.
Collapse
Affiliation(s)
- Erica M Pasini
- Department of Parasitology, Biomedical Primate Research Center, Rijswijk, Netherlands
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Center, Rijswijk, Netherlands
| |
Collapse
|
18
|
Voorberg-van der Wel A, Kocken CHM, Zeeman AM. Modeling Relapsing Malaria: Emerging Technologies to Study Parasite-Host Interactions in the Liver. Front Cell Infect Microbiol 2021; 10:606033. [PMID: 33585277 PMCID: PMC7878928 DOI: 10.3389/fcimb.2020.606033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/04/2020] [Indexed: 01/03/2023] Open
Abstract
Recent studies of liver stage malaria parasite-host interactions have provided exciting new insights on the cross-talk between parasite and its mammalian (predominantly rodent) host. We review the latest state of the art and and zoom in on new technologies that will provide the tools necessary to investigate host-parasite interactions of relapsing parasites. Interactions between hypnozoites and hepatocytes are particularly interesting because the parasite can remain in a quiescent state for prolonged periods of time and triggers for reactivation have not been irrefutably identified. If we learn more about the cross-talk between hypnozoite and host we may be able to identify factors that encourage waking up these dormant parasite reservoirs and help to achieve the total eradication of malaria.
Collapse
Affiliation(s)
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Center, Rijswijk, Netherlands
| | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Center, Rijswijk, Netherlands
| |
Collapse
|
19
|
Galinski MR. Functional genomics of simian malaria parasites and host-parasite interactions. Brief Funct Genomics 2020; 18:270-280. [PMID: 31241151 PMCID: PMC6859816 DOI: 10.1093/bfgp/elz013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/21/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022] Open
Abstract
Two simian malaria parasite species, Plasmodium knowlesi and Plasmodium cynomolgi, cause zoonotic infections in Southeast Asia, and they have therefore gained recognition among scientists and public health officials. Notwithstanding, these species and others including Plasmodium coatneyi have served for decades as sources of knowledge on the biology, genetics and evolution of Plasmodium, and the diverse ramifications and outcomes of malaria in their monkey hosts. Experimental analysis of these species can help to fill gaps in knowledge beyond what may be possible studying the human malaria parasites or rodent parasite species. The genome sequences for these simian malaria parasite species were reported during the last decade, and functional genomics research has since been pursued. Here research on the functional genomics analysis involving these species is summarized and their importance is stressed, particularly for understanding host–parasite interactions, and potentially testing novel interventions. Importantly, while Plasmodium falciparum and Plasmodium vivax can be studied in small New World monkeys, the simian malaria parasites can be studied more effectively in the larger Old World monkey macaque hosts, which are more closely related to humans. In addition to ex vivo analyses, experimental scenarios can include passage through Anopheline mosquito hosts and longitudinal infections in monkeys to study acute and chronic infections, as well as relapses, all in the context of the in vivo host environment. Such experiments provide opportunities for understanding functional genomic elements that govern host–parasite interactions, immunity and pathogenesis in-depth, addressing hypotheses not possible from in vitro cultures or cross-sectional clinical studies with humans.
Collapse
Affiliation(s)
- Mary R Galinski
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
20
|
Rogers KJ, Shtanko O, Vijay R, Mallinger LN, Joyner CJ, Galinski MR, Butler NS, Maury W. Acute Plasmodium Infection Promotes Interferon-Gamma-Dependent Resistance to Ebola Virus Infection. Cell Rep 2020; 30:4041-4051.e4. [PMID: 32209467 PMCID: PMC7172281 DOI: 10.1016/j.celrep.2020.02.104] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/06/2020] [Accepted: 02/27/2020] [Indexed: 10/24/2022] Open
Abstract
During the 2013-2016 Ebola virus (EBOV) epidemic, a significant number of patients admitted to Ebola treatment units were co-infected with Plasmodium falciparum, a predominant agent of malaria. However, there is no consensus on how malaria impacts EBOV infection. The effect of acute Plasmodium infection on EBOV challenge was investigated using mouse-adapted EBOV and a biosafety level 2 (BSL-2) model virus. We demonstrate that acute Plasmodium infection protects from lethal viral challenge, dependent upon interferon gamma (IFN-γ) elicited as a result of parasite infection. Plasmodium-infected mice lacking the IFN-γ receptor are not protected. Ex vivo incubation of naive human or mouse macrophages with sera from acutely parasitemic rodents or macaques programs a proinflammatory phenotype dependent on IFN-γ and renders cells resistant to EBOV infection. We conclude that acute Plasmodium infection can safeguard against EBOV by the production of protective IFN-γ. These findings have implications for anti-malaria therapies administered during episodic EBOV outbreaks in Africa.
Collapse
Affiliation(s)
- Kai J Rogers
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Olena Shtanko
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Rahul Vijay
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Laura N Mallinger
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Chester J Joyner
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA; Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Center, Emory University, Atlanta, GA 30322, USA
| | - Mary R Galinski
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Center, Emory University, Atlanta, GA 30322, USA; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Noah S Butler
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
21
|
A dual fluorescent Plasmodium cynomolgi reporter line reveals in vitro malaria hypnozoite reactivation. Commun Biol 2020; 3:7. [PMID: 31909199 PMCID: PMC6941962 DOI: 10.1038/s42003-019-0737-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022] Open
Abstract
Plasmodium vivax malaria is characterized by repeated episodes of blood stage infection (relapses) resulting from activation of dormant stages in the liver, so-called hypnozoites. Transition of hypnozoites into developing schizonts has never been observed. A barrier for studying this has been the lack of a system in which to monitor growth of liver stages. Here, exploiting the unique strengths of the simian hypnozoite model P. cynomolgi, we have developed green-fluorescent (GFP) hypnozoites that turn on red-fluorescent (mCherry) upon activation. The transgenic parasites show full liver stage development, including merozoite release and red blood cell infection. We demonstrate that individual hypnozoites actually can activate and resume development after prolonged culture, providing the last missing evidence of the hypnozoite theory of relapse. The few events identified indicate that hypnozoite activation in vitro is infrequent. This system will further our understanding of the mechanisms of hypnozoite activation and may facilitate drug discovery approaches.
Collapse
|
22
|
Liu G, Li Y, Qin L, Yan Y, Ye Y, Chen Y, Huang C, Zhao S, Yao Y, Su Z, Chen X. SIV infection aggravates malaria in a Chinese rhesus monkey coinfection model. BMC Infect Dis 2019; 19:965. [PMID: 31718574 PMCID: PMC6852750 DOI: 10.1186/s12879-019-4465-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 09/11/2019] [Indexed: 01/26/2023] Open
Abstract
Background The co-occurrence of human immunodeficiency virus (HIV) infection and malaria in humans in endemic areas raises the question of whether one of these infections affects the course of the other. Although epidemiological studies have shown the impact of HIV infection on malaria, the mechanism(s) are not yet fully understood. Using a Chinese rhesus macaque coinfection model with simian immunodeficiency virus (SIV) and Plasmodium cynomolgi (Pc) malaria, we investigated the effect of concurrent SIV infection on the course of malaria and the underlying immunological mechanism(s). Methods We randomly assigned ten Chinese rhesus monkeys to two groups based on body weight and age. The SIV-Pc coinfection animals (S + P group) were infected intravenously with SIVmac251 eight weeks prior to malaria infection, and the control animals (P group) were infected intravenously with only Pc-infected red blood cells. After malaria was cured with chloroquine phosphate, we also initiated a secondary malaria infection that lasted 4 weeks. We monitored body weight, body temperature and parasitemia, measured SIV viral loads, hemoglobin and neopterin, and tracked the CD4+, CD8+, and CD4+ memory subpopulations, Ki67 and apoptosis by flow cytometry. Then, we compared these parameters between the two groups. Results The animals infected with SIV prior to Pc infection exhibited more severe malaria symptoms characterized by longer episodes, higher parasitemia, more severe anemia, greater body weight loss and higher body temperature than the animals infected with Pc alone. Concurrent SIV infection also impaired immune protection against the secondary Pc challenge infection. The coinfected animals showed a reduced B cell response to Pc malaria and produced lower levels of Pc-specific antibodies. In addition, compared to the animals subjected to Pc infection alone, the animals coinfected with SIV and Pc had suppressed total CD4+ T cells, CD4+CD28highCD95high central memory T cells, and CD4+CD28lowCD95− naïve T cells, which may result from the imbalanced immune activation and faster CD4+ T cell turnover in coinfected animals. Conclusions SIV infection aggravates malaria physiologically and immunologically in Chinese rhesus monkeys. This nonhuman primate SIV and Pc malaria coinfection model might be a useful tool for investigating human HIV and malaria coinfection and developing effective therapeutics.
Collapse
Affiliation(s)
- Guangjie Liu
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Diseases, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou, 510530, China.,Graduate School, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.,Shenzhen Institute of Geriatrics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Youjia Li
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Diseases, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou, 510530, China.,Shenzhen Institute of Geriatrics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Li Qin
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Diseases, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou, 510530, China
| | - Yongxiang Yan
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Diseases, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou, 510530, China
| | - Yijian Ye
- Laboratory of Immunobiology, State Key Laboratory of Respiratory Disease, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yue Chen
- Laboratory of Immunobiology, State Key Laboratory of Respiratory Disease, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Cuizhu Huang
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Diseases, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou, 510530, China
| | - Siting Zhao
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Diseases, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou, 510530, China
| | - Yongchao Yao
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Diseases, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou, 510530, China.,Shenzhen Institute of Geriatrics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Su
- Laboratory of Immunobiology, State Key Laboratory of Respiratory Disease, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Xiaoping Chen
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Diseases, Center of Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Guangzhou, 510530, China.
| |
Collapse
|
23
|
Caldelari R, Dogga S, Schmid MW, Franke-Fayard B, Janse CJ, Soldati-Favre D, Heussler V. Transcriptome analysis of Plasmodium berghei during exo-erythrocytic development. Malar J 2019; 18:330. [PMID: 31551073 PMCID: PMC6760107 DOI: 10.1186/s12936-019-2968-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The complex life cycle of malaria parasites requires well-orchestrated stage specific gene expression. In the vertebrate host the parasites grow and multiply by schizogony in two different environments: within erythrocytes and within hepatocytes. Whereas erythrocytic parasites are well-studied in this respect, relatively little is known about the exo-erythrocytic stages. METHODS In an attempt to fill this gap, genome wide RNA-seq analyses of various exo-erythrocytic stages of Plasmodium berghei including sporozoites, samples from a time-course of liver stage development and detached cells were performed. These latter contain infectious merozoites and represent the final step in exo-erythrocytic development. RESULTS The analysis represents the complete transcriptome of the entire life cycle of P. berghei parasites with temporal detailed analysis of the liver stage allowing comparison of gene expression across the progression of the life cycle. These RNA-seq data from different developmental stages were used to cluster genes with similar expression profiles, in order to infer their functions. A comparison with published data from other parasite stages confirmed stage-specific gene expression and revealed numerous genes that are expressed differentially in blood and exo-erythrocytic stages. One of the most exo-erythrocytic stage-specific genes was PBANKA_1003900, which has previously been annotated as a "gametocyte specific protein". The promoter of this gene drove high GFP expression in exo-erythrocytic stages, confirming its expression profile seen by RNA-seq. CONCLUSIONS The comparative analysis of the genome wide mRNA expression profiles of erythrocytic and different exo-erythrocytic stages could be used to improve the understanding of gene regulation in Plasmodium parasites and can be used to model exo-erythrocytic stage metabolic networks toward the identification of differences in metabolic processes during schizogony in erythrocytes and hepatocytes.
Collapse
Affiliation(s)
- Reto Caldelari
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| | - Sunil Dogga
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva CMU, Geneva, Switzerland
| | | | - Blandine Franke-Fayard
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Chris J Janse
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva CMU, Geneva, Switzerland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| |
Collapse
|
24
|
Humoral immunity prevents clinical malaria during Plasmodium relapses without eliminating gametocytes. PLoS Pathog 2019; 15:e1007974. [PMID: 31536608 PMCID: PMC6752766 DOI: 10.1371/journal.ppat.1007974] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/10/2019] [Indexed: 12/19/2022] Open
Abstract
Plasmodium relapses are attributed to the activation of dormant liver-stage parasites and are responsible for a significant number of recurring malaria blood-stage infections. While characteristic of human infections caused by P. vivax and P. ovale, their relative contribution to malaria disease burden and transmission remains poorly understood. This is largely because it is difficult to identify ‘bona fide’ relapse infections due to ongoing transmission in most endemic areas. Here, we use the P. cynomolgi–rhesus macaque model of relapsing malaria to demonstrate that clinical immunity can form after a single sporozoite-initiated blood-stage infection and prevent illness during relapses and homologous reinfections. By integrating data from whole blood RNA-sequencing, flow cytometry, P. cynomolgi-specific ELISAs, and opsonic phagocytosis assays, we demonstrate that this immunity is associated with a rapid recall response by memory B cells that expand and produce anti-parasite IgG1 that can mediate parasite clearance of relapsing parasites. The reduction in parasitemia during relapses was mirrored by a reduction in the total number of circulating gametocytes, but importantly, the cumulative proportion of gametocytes increased during relapses. Overall, this study reveals that P. cynomolgi relapse infections can be clinically silent in macaques due to rapid memory B cell responses that help to clear asexual-stage parasites but still carry gametocytes. Plasmodium vivax contributes significantly to global malaria morbidity and remains a major obstacle for malaria elimination due to its ability to form dormant stages in the liver. These forms can become activated to cause relapsing blood-stage infections. Relapses remain poorly understood because it is difficult to verify whether P. vivax blood-stage infections in patients are due to new infections or relapses in most cases. Here, we use a nonhuman primate model of Plasmodium vivax malaria in concert with state-of-the-art immunological and molecular techniques to assess pathogenesis, host responses, and circulating gametocyte levels during relapses. We found that relapses were clinically silent compared to initial infections, and they were associated with a robust memory B cell response. This response resulted in the production of antibodies that were able to mediate clearance of asexual parasites. Despite this rapid immune protection, the sexual-stage gametocytes continued to circulate. Our study provides mechanistic insights into the host-parasite interface during Plasmodium relapse infections and demonstrates that clinically silent relapses can harbor gametocytes that may be infectious to mosquitoes.
Collapse
|
25
|
Cordy RJ, Patrapuvich R, Lili LN, Cabrera-Mora M, Chien JT, Tharp GK, Khadka M, Meyer EV, Lapp SA, Joyner CJ, Garcia A, Banton S, Tran V, Luvira V, Rungin S, Saeseu T, Rachaphaew N, Pakala SB, DeBarry JD, Kissinger JC, Ortlund EA, Bosinger SE, Barnwell JW, Jones DP, Uppal K, Li S, Sattabongkot J, Moreno A, Galinski MR. Distinct amino acid and lipid perturbations characterize acute versus chronic malaria. JCI Insight 2019; 4:125156. [PMID: 31045574 DOI: 10.1172/jci.insight.125156] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
Abstract
Chronic malaria is a major public health problem and significant challenge for disease eradication efforts. Despite its importance, the biological factors underpinning chronic malaria are not fully understood. Recent studies have shown that host metabolic state can influence malaria pathogenesis and transmission, but its role in chronicity is not known. Here, with the goal of identifying distinct modifications in the metabolite profiles of acute versus chronic malaria, metabolomics was performed on plasma from Plasmodium-infected humans and nonhuman primates with a range of parasitemias and clinical signs. In rhesus macaques infected with Plasmodium coatneyi, significant alterations in amines, carnitines, and lipids were detected during a high parasitemic acute phase and many of these reverted to baseline levels once a low parasitemic chronic phase was established. Plasmodium gene expression, studied in parallel in the macaques, revealed transcriptional changes in amine, fatty acid, lipid and energy metabolism genes, as well as variant antigen genes. Furthermore, a common set of amines, carnitines, and lipids distinguished acute from chronic malaria in plasma from human Plasmodium falciparum cases. In summary, distinct host-parasite metabolic environments have been uncovered that characterize acute versus chronic malaria, providing insights into the underlying host-parasite biology of malaria disease progression.
Collapse
Affiliation(s)
- Regina Joice Cordy
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Biology, Wake Forest University, Winston-Salem, North Carolina, USA
| | | | - Loukia N Lili
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA.,Department of Genetics and Genomic Sciences, Institute for Next Generation Healthcare, Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | - Monica Cabrera-Mora
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Jung-Ting Chien
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Gregory K Tharp
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Manoj Khadka
- Emory Integrated Lipidomics Core, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Esmeralda Vs Meyer
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Stacey A Lapp
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Chester J Joyner
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - AnaPatricia Garcia
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Sophia Banton
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - ViLinh Tran
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Viravarn Luvira
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Siriwan Rungin
- Mahidol Vivax Research Unit, Mahidol University, Bangkok, Thailand
| | - Teerawat Saeseu
- Mahidol Vivax Research Unit, Mahidol University, Bangkok, Thailand
| | | | | | | | | | - Jessica C Kissinger
- Institute of Bioinformatics.,Center for Tropical and Emerging Global Diseases, and.,Department of Genetics, University of Georgia, Athens, Georgia, USA
| | - Eric A Ortlund
- Emory Integrated Lipidomics Core, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Steven E Bosinger
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - John W Barnwell
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Karan Uppal
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Shuzhao Li
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | | | - Alberto Moreno
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Mary R Galinski
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
26
|
Taghavian O, Jain A, Joyner CJ, Ketchum S, Nakajima R, Jasinskas A, Liang L, Fong R, King C, Greenhouse B, Murphy M, Bailey J, Galinski MR, Barnwell JW, Plowe CV, Davies DH, Felgner PL. Antibody Profiling by Proteome Microarray with Multiplex Isotype Detection Reveals Overlap between Human and Aotus nancymaae Controlled Malaria Infections. Proteomics 2019; 18. [PMID: 29266845 DOI: 10.1002/pmic.201700277] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/21/2017] [Indexed: 12/13/2022]
Abstract
The development of vaccines against malaria and serodiagnostic tests for detecting recent exposure requires tools for antigen discovery and suitable animal models. The protein microarray is a high-throughput, sample sparing technique, with applications in infectious disease research, clinical diagnostics, epidemiology, and vaccine development. We recently demonstrated Qdot-based indirect immunofluorescence together with portable optical imager ArrayCAM using single isotype detection could replicate data using the conventional laser confocal scanner system. We developed a multiplexing protocol for simultaneous detection of IgG, IgA, and IgM and compared samples from a controlled human malaria infection model with those from controlled malaria infections of Aotus nancymaae, a widely used non-human primate model of human malaria. IgG profiles showed the highest concordance in number of reactive antigens; thus, of the 139 antigens recognized by human IgG antibody, 111 were also recognized by Aotus monkeys. Interestingly, IgA profiles were largely non-overlapping. Finally, on the path toward wider deployment of the portable platform, we show excellent correlations between array data obtained in five independent laboratories around the United States using the multiplexing protocol (R2 : 0.60-0.92). This study supports the use of this platform for wider deployment, particularly in endemic areas where such a tool will have the greatest impact on global human health.
Collapse
Affiliation(s)
- Omid Taghavian
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Aarti Jain
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Chester J Joyner
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | - Rie Nakajima
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Algis Jasinskas
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Li Liang
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Rich Fong
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Christopher King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Bryan Greenhouse
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Maxwell Murphy
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Jason Bailey
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Mary R Galinski
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - John W Barnwell
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Christopher V Plowe
- Howard Hughes Medical Institute/Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - D Huw Davies
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Philip L Felgner
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
27
|
Fonseca LL, Joyner CJ, Saney CL, Moreno A, Barnwell JW, Galinski MR, Voit EO. Analysis of erythrocyte dynamics in Rhesus macaque monkeys during infection with Plasmodium cynomolgi. Malar J 2018; 17:410. [PMID: 30400896 PMCID: PMC6219197 DOI: 10.1186/s12936-018-2560-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 10/30/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Malaria is a major mosquito transmitted, blood-borne parasitic disease that afflicts humans. The disease causes anaemia and other clinical complications, which can lead to death. Plasmodium vivax is known for its reticulocyte host cell specificity, but many gaps in disease details remain. Much less is known about the closely related species, Plasmodium cynomolgi, although it is naturally acquired and causes zoonotic malaria. Here, a computational model is developed based on longitudinal analyses of P. cynomolgi infections in nonhuman primates to investigate the erythrocyte dynamics that is pertinent to understanding both P. cynomolgi and P. vivax malaria in humans. METHODS A cohort of five P. cynomolgi infected Rhesus macaques (Macaca mulatta) is studied, with individuals exhibiting a plethora of clinical outcomes, including varying levels of anaemia. A discrete recursive model with age structure is developed to replicate the dynamics of P. cynomolgi blood-stage infections. The model allows for parasitic reticulocyte preference and assumes an age preference among the mature RBCs. RBC senescence is modelled using a hazard function, according to which RBCs have a mean lifespan of 98 ± 21 days. RESULTS Based on in vivo data from three cohorts of macaques, the computational model is used to characterize the reticulocyte lifespan in circulation as 24 ± 5 h (n = 15) and the rate of RBC production as 2727 ± 209 cells/h/µL (n = 15). Analysis of the host responses reveals a pre-patency increase in the number of reticulocytes. It also allows the quantification of RBC removal through the bystander effect. CONCLUSIONS The evident pre-patency increase in reticulocytes is due to a shift towards the release of younger reticulocytes, which could result from a parasite-induced factor meant to increase reticulocyte availability and satisfy the parasite's tropism, which has an average value of 32:1 in this cohort. The number of RBCs lost due to the bystander effect relative to infection-induced RBC losses is 62% for P. cynomolgi infections, which is substantially lower than the value of 95% previously determined for another simian species, Plasmodium coatneyi.
Collapse
Affiliation(s)
- Luis L Fonseca
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332-2000, USA.
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30322, USA.
| | - Chester J Joyner
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30322, USA
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Celia L Saney
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30322, USA
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Alberto Moreno
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30322, USA
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - John W Barnwell
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30322, USA
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, 30322, USA
| | - Mary R Galinski
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30322, USA
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Eberhard O Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332-2000, USA
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
28
|
McBirney SE, Chen D, Scholtz A, Ameri H, Armani AM. Rapid Diagnostic for Point-of-Care Malaria Screening. ACS Sens 2018; 3:1264-1270. [PMID: 29781606 DOI: 10.1021/acssensors.8b00269] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Despite significant success in therapeutic development, malaria remains a widespread and deadly infectious disease in the developing world. Given the nearly 100% efficacy of current malaria therapeutics, the primary barrier to eradication is lack of early diagnosis of the infected population. However, there are multiple strains of malaria. Although significant efforts and resources have been invested in developing antibody-based diagnostic methods for Plasmodium falciparum, a rapid and easy to use screening method capable of detecting all malaria strains has not been realized. Yet, until the entire malaria-infected population receives treatment, the disease will continue to impact society. Here, we report the development of a portable, magneto-optic technology for early stage malaria diagnosis based on the detection of the malaria pigment, hemozoin. Using β-hematin, a hemozoin mimic, we demonstrate detection limits of <0.0081 μg/mL in 500 μL of whole rabbit blood with no additional reagents required. This level corresponds to <26 parasites/μL, a full order of magnitude below clinical relevance and comparable to or less than existing technologies.
Collapse
Affiliation(s)
| | | | - Alexis Scholtz
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Hossein Ameri
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California 90033, United States
| | | |
Collapse
|
29
|
Plasmodium vivax and Plasmodium falciparum infection dynamics: re-infections, recrudescences and relapses. Malar J 2018; 17:170. [PMID: 29665803 PMCID: PMC5905131 DOI: 10.1186/s12936-018-2318-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022] Open
Abstract
Background In malaria endemic populations, complex patterns of Plasmodium vivax and Plasmodium falciparum blood-stage infection dynamics may be observed. Genotyping samples from longitudinal cohort studies for merozoite surface protein (msp) variants increases the information available in the data, allowing multiple infecting parasite clones in a single individual to be identified. msp genotyped samples from two longitudinal cohorts in Papua New Guinea (PNG) and Thailand were analysed using a statistical model where the times of acquisition and clearance of each clone in every individual were estimated using a process of data augmentation. Results For the populations analysed, the duration of blood-stage P. falciparum infection was estimated as 36 (95% Credible Interval (CrI): 29, 44) days in PNG, and 135 (95% CrI 94, 191) days in Thailand. Experiments on simulated data indicated that it was not possible to accurately estimate the duration of blood-stage P. vivax infections due to the lack of identifiability between a single blood-stage infection and multiple, sequential blood-stage infections caused by relapses. Despite this limitation, the method and data point towards short duration of blood-stage P. vivax infection with a lower bound of 24 days in PNG, and 29 days in Thailand. On an individual level, P. vivax recurrences cannot be definitively classified into re-infections, recrudescences or relapses, but a probabilistic relapse phenotype can be assigned to each P. vivax sample, allowing investigation of the association between epidemiological covariates and the incidence of relapses. Conclusion The statistical model developed here provides a useful new tool for in-depth analysis of malaria data from longitudinal cohort studies, and future application to data sets with multi-locus genotyping will allow more detailed investigation of infection dynamics. Electronic supplementary material The online version of this article (10.1186/s12936-018-2318-1) contains supplementary material, which is available to authorized users.
Collapse
|
30
|
Galinski MR, Lapp SA, Peterson MS, Ay F, Joyner CJ, LE Roch KG, Fonseca LL, Voit EO. Plasmodium knowlesi: a superb in vivo nonhuman primate model of antigenic variation in malaria. Parasitology 2018; 145:85-100. [PMID: 28712361 PMCID: PMC5798396 DOI: 10.1017/s0031182017001135] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/03/2017] [Accepted: 06/06/2017] [Indexed: 02/08/2023]
Abstract
Antigenic variation in malaria was discovered in Plasmodium knowlesi studies involving longitudinal infections of rhesus macaques (M. mulatta). The variant proteins, known as the P. knowlesi Schizont Infected Cell Agglutination (SICA) antigens and the P. falciparum Erythrocyte Membrane Protein 1 (PfEMP1) antigens, expressed by the SICAvar and var multigene families, respectively, have been studied for over 30 years. Expression of the SICA antigens in P. knowlesi requires a splenic component, and specific antibodies are necessary for variant antigen switch events in vivo. Outstanding questions revolve around the role of the spleen and the mechanisms by which the expression of these variant antigen families are regulated. Importantly, the longitudinal dynamics and molecular mechanisms that govern variant antigen expression can be studied with P. knowlesi infection of its mammalian and vector hosts. Synchronous infections can be initiated with established clones and studied at multi-omic levels, with the benefit of computational tools from systems biology that permit the integration of datasets and the design of explanatory, predictive mathematical models. Here we provide an historical account of this topic, while highlighting the potential for maximizing the use of P. knowlesi - macaque model systems and summarizing exciting new progress in this area of research.
Collapse
Affiliation(s)
- M R Galinski
- Emory Vaccine Center,Yerkes National Primate Research Center,Emory University,Atlanta,GA,USA
| | - S A Lapp
- Emory Vaccine Center,Yerkes National Primate Research Center,Emory University,Atlanta,GA,USA
| | - M S Peterson
- Emory Vaccine Center,Yerkes National Primate Research Center,Emory University,Atlanta,GA,USA
| | - F Ay
- La Jolla Institute for Allergy and Immunology,La Jolla,CA 92037,USA
| | - C J Joyner
- Emory Vaccine Center,Yerkes National Primate Research Center,Emory University,Atlanta,GA,USA
| | - K G LE Roch
- Department of Cell Biology & Neuroscience,Center for Disease and Vector Research,Institute for Integrative Genome Biology,University of California Riverside,CA 92521,USA
| | - L L Fonseca
- The Wallace H. Coulter Department of Biomedical Engineering,Georgia Institute of Technology and Emory University,Atlanta,Georgia,30332-2000,USA
| | - E O Voit
- The Wallace H. Coulter Department of Biomedical Engineering,Georgia Institute of Technology and Emory University,Atlanta,Georgia,30332-2000,USA
| |
Collapse
|
31
|
Gardinassi LG, Cordy RJ, Lacerda MVG, Salinas JL, Monteiro WM, Melo GC, Siqueira AM, Val FF, Tran V, Jones DP, Galinski MR, Li S. Metabolome-wide association study of peripheral parasitemia in Plasmodium vivax malaria. Int J Med Microbiol 2017; 307:533-541. [PMID: 28927849 PMCID: PMC5698147 DOI: 10.1016/j.ijmm.2017.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/26/2017] [Accepted: 09/03/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Plasmodium vivax is one of the leading causes of malaria worldwide. Infections with this parasite cause diverse clinical manifestations, and recent studies revealed that infections with P. vivax can result in severe and fatal disease. Despite these facts, biological traits of the host response and parasite metabolism during P. vivax malaria are still largely underexplored. Parasitemia is clearly related to progression and severity of malaria caused by P. falciparum, however the effects of parasitemia during infections with P. vivax are not well understood. RESULTS We conducted an exploratory study using a high-resolution metabolomics platform that uncovered significant associations between parasitemia levels and plasma metabolites from 150 patients with P. vivax malaria. Most plasma metabolites were inversely associated with higher levels of parasitemia. Top predicted metabolites are implicated into pathways of heme and lipid metabolism, which include biliverdin, bilirubin, palmitoylcarnitine, stearoylcarnitine, phosphocholine, glycerophosphocholine, oleic acid and omega-carboxy-trinor-leukotriene B4. CONCLUSIONS The abundance of several plasma metabolites varies according to the levels of parasitemia in patients with P. vivax malaria. Moreover, our data suggest that the host response and/or parasite survival might be affected by metabolites involved in the degradation of heme and metabolism of several lipids. Importantly, these data highlight metabolic pathways that may serve as targets for the development of new antimalarial compounds.
Collapse
Affiliation(s)
- Luiz Gustavo Gardinassi
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Malaria Host-Pathogen Interaction Center, Atlanta, GA, USA
| | - Regina Joice Cordy
- Malaria Host-Pathogen Interaction Center, Atlanta, GA, USA; International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Marcus V G Lacerda
- Gerência de Malária, Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM, Brazil; Escola Superior de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, AM, Brazil; Instituto Leônidas & Maria Deane (FIOCRUZ), Manaus, AM, Brazil
| | | | - Wuelton M Monteiro
- Gerência de Malária, Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM, Brazil; Escola Superior de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, AM, Brazil
| | - Gisely C Melo
- Gerência de Malária, Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM, Brazil; Escola Superior de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, AM, Brazil
| | - André M Siqueira
- Instituto Nacional de Infectologia Evandro Chagas (FIOCRUZ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando F Val
- Gerência de Malária, Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM, Brazil; Escola Superior de Ciências da Saúde, Universidade do Estado do Amazonas, Manaus, AM, Brazil
| | - ViLinh Tran
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Malaria Host-Pathogen Interaction Center, Atlanta, GA, USA; Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Malaria Host-Pathogen Interaction Center, Atlanta, GA, USA; Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Mary R Galinski
- Malaria Host-Pathogen Interaction Center, Atlanta, GA, USA; International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA; Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Shuzhao Li
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA; Malaria Host-Pathogen Interaction Center, Atlanta, GA, USA; Clinical Biomarkers Laboratory, Department of Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
32
|
Tang Y, Gupta A, Garimalla S, Galinski MR, Styczynski MP, Fonseca LL, Voit EO. Metabolic modeling helps interpret transcriptomic changes during malaria. Biochim Biophys Acta Mol Basis Dis 2017; 1864:2329-2340. [PMID: 29069611 DOI: 10.1016/j.bbadis.2017.10.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 09/27/2017] [Accepted: 10/17/2017] [Indexed: 10/18/2022]
Abstract
Disease represents a specific case of malfunctioning within a complex system. Whereas it is often feasible to observe and possibly treat the symptoms of a disease, it is much more challenging to identify and characterize its molecular root causes. Even in infectious diseases that are caused by a known parasite, it is often impossible to pinpoint exactly which molecular profiles of components or processes are directly or indirectly altered. However, a deep understanding of such profiles is a prerequisite for rational, efficacious treatments. Modern omics methodologies are permitting large-scale scans of some molecular profiles, but these scans often yield results that are not intuitive and difficult to interpret. For instance, the comparison of healthy and diseased transcriptome profiles may point to certain sets of involved genes, but a host of post-transcriptional processes and regulatory mechanisms renders predictions regarding metabolic or physiological consequences of the observed changes in gene expression unreliable. Here we present proof of concept that dynamic models of metabolic pathway systems may offer a tool for interpreting transcriptomic profiles measured during disease. We illustrate this strategy with the interpretation of expression data of genes coding for enzymes associated with purine metabolism. These data were obtained during infections of rhesus macaques (Macaca mulatta) with the malaria parasite Plasmodium cynomolgi or P. coatneyi. The model-based interpretation reveals clear patterns of flux redistribution within the purine pathway that are consistent between the two malaria pathogens and are even reflected in data from humans infected with P. falciparum. This article is part of a Special Issue entitled: Accelerating Precision Medicine through Genetic and Genomic Big Data Analysis edited by Yudong Cai & Tao Huang.
Collapse
Affiliation(s)
- Yan Tang
- School of Chemical and Biomolecular Engineering, Georgia Tech, Atlanta, GA 30332, USA
| | - Anuj Gupta
- Department of Biomedical Engineering, Georgia Tech, Atlanta, GA 30332, USA
| | - Swetha Garimalla
- School of Biological Sciences, Georgia Tech, Atlanta, GA 30332, USA
| | -
- Malaria Host-Pathogen Interaction Center, USA
| | - Mary R Galinski
- Emory Vaccine Center at Yerkes, Emory University, 954 Gatewood Road, EVC 003, Atlanta, GA 30329, USA; Department of Medicine, Division of Infectious Diseases, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Mark P Styczynski
- School of Chemical and Biomolecular Engineering, Georgia Tech, Atlanta, GA 30332, USA
| | - Luis L Fonseca
- Department of Biomedical Engineering, Georgia Tech, Atlanta, GA 30332, USA
| | - Eberhard O Voit
- Department of Biomedical Engineering, Georgia Tech, Atlanta, GA 30332, USA.
| |
Collapse
|
33
|
Tang Y, Joyner CJ, Cabrera-Mora M, Saney CL, Lapp SA, Nural MV, Pakala SB, DeBarry JD, Soderberg S, Kissinger JC, Lamb TJ, Galinski MR, Styczynski MP. Integrative analysis associates monocytes with insufficient erythropoiesis during acute Plasmodium cynomolgi malaria in rhesus macaques. Malar J 2017; 16:384. [PMID: 28938907 PMCID: PMC5610412 DOI: 10.1186/s12936-017-2029-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/12/2017] [Indexed: 01/06/2023] Open
Abstract
Background Mild to severe anaemia is a common complication of malaria that is caused in part by insufficient erythropoiesis in the bone marrow. This study used systems biology to evaluate the transcriptional and alterations in cell populations in the bone marrow during Plasmodium cynomolgi infection of rhesus macaques (a model of Plasmodium vivax malaria) that may affect erythropoiesis. Results An appropriate erythropoietic response did not occur to compensate for anaemia during acute cynomolgi malaria despite an increase in erythropoietin levels. During this period, there were significant perturbations in the bone marrow transcriptome. In contrast, relapses did not induce anaemia and minimal changes in the bone marrow transcriptome were detected. The differentially expressed genes during acute infection were primarily related to ongoing inflammatory responses with significant contributions from Type I and Type II Interferon transcriptional signatures. These were associated with increased frequency of intermediate and non-classical monocytes. Recruitment and/or expansion of these populations was correlated with a decrease in the erythroid progenitor population during acute infection, suggesting that monocyte-associated inflammation may have contributed to anaemia. The decrease in erythroid progenitors was associated with downregulation of genes regulated by GATA1 and GATA2, two master regulators of erythropoiesis, providing a potential molecular basis for these findings. Conclusions These data suggest the possibility that malarial anaemia may be driven by monocyte-associated disruption of GATA1/GATA2 function in erythroid progenitors resulting in insufficient erythropoiesis during acute infection. Electronic supplementary material The online version of this article (doi:10.1186/s12936-017-2029-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Tang
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Chester J Joyner
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Monica Cabrera-Mora
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Celia L Saney
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Stacey A Lapp
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Mustafa V Nural
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Institute of Bioinformatics, University of Georgia, Athens, GA, USA.,Department of Computer Science, University of Georgia, Athens, GA, USA
| | - Suman B Pakala
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| | - Jeremy D DeBarry
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| | - Stephanie Soderberg
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | - Jessica C Kissinger
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Institute of Bioinformatics, University of Georgia, Athens, GA, USA.,Department of Genetics, University of Georgia, Athens, GA, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA.,Department of Computer Science, University of Georgia, Athens, GA, USA
| | - Tracey J Lamb
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Mary R Galinski
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Mark P Styczynski
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA. .,Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
| |
Collapse
|
34
|
Fonseca LL, Joyner CJ, Galinski MR, Voit EO. A model of Plasmodium vivax concealment based on Plasmodium cynomolgi infections in Macaca mulatta. Malar J 2017; 16:375. [PMID: 28923058 PMCID: PMC5608162 DOI: 10.1186/s12936-017-2008-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 09/02/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Plasmodium vivax can cause severe malaria. The total parasite biomass during infections is correlated with the severity of disease but not necessarily quantified accurately by microscopy. This finding has raised the question whether there could be sub-populations of parasites that are not observed in peripheral blood smears but continue to contribute to the increase in parasite numbers that drive pathogenesis. Non-human primate infection models utilizing the closely related simian malaria parasite Plasmodium cynomolgi hold the potential for quantifying the magnitude of possibly unobserved infected red blood cell (iRBC) populations and determining how the presence of this hidden reservoir correlates with disease severity. METHODS Time series data tracking the longitudinal development of parasitaemia in five Macaca mulatta infected with P. cynomolgi were used to design a computational model quantifying iRBCs that circulate in the blood versus those that are not detectable and are termed here as 'concealed'. This terminology is proposed to distinguish such observations from the deep vascular and widespread 'sequestration' of Plasmodium falciparum iRBCs, which is governed by distinctly different molecular mechanisms. RESULTS The computational model presented here clearly demonstrates that the observed growth data of iRBC populations are not consistent with the known biology and blood-stage cycle of P. cynomolgi. However, the discrepancies can be resolved when a sub-population of concealed iRBCs is taken into account. The model suggests that the early growth of a hidden parasite sub-population has the potential to drive disease. As an alternative, the data could be explained by the sequential release of merozoites from the liver over a number of days, but this scenario seems less likely. CONCLUSIONS Concealment of a non-circulating iRBC sub-population during P. cynomolgi infection of M. mulatta is an important aspect of this successful host-pathogen relationship. The data also support the likelihood that a sub-population of iRBCs of P. vivax has a comparable means to become withdrawn from the peripheral circulation. This inference has implications for understanding vivax biology and pathogenesis and stresses the importance of considering a concealed parasite reservoir with regard to vivax epidemiology and the quantification and treatment of P. vivax infections.
Collapse
Affiliation(s)
- Luis L Fonseca
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Drive, Suite 2115, Atlanta, GA, 30332-2000, USA.,Malaria Host-Pathogen Interaction Center, Atlanta, GA, USA
| | - Chester J Joyner
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA.,Malaria Host-Pathogen Interaction Center, Atlanta, GA, USA
| | | | - Mary R Galinski
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, 954 Gatewood Road, Atlanta, GA, 30329, USA.,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA.,Malaria Host-Pathogen Interaction Center, Atlanta, GA, USA
| | - Eberhard O Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Drive, Suite 2115, Atlanta, GA, 30332-2000, USA. .,Malaria Host-Pathogen Interaction Center, Atlanta, GA, USA.
| |
Collapse
|
35
|
J Joyner C, Consortium TM, Wood JS, Moreno A, Garcia A, Galinski MR. Case Report: Severe and Complicated Cynomolgi Malaria in a Rhesus Macaque Resulted in Similar Histopathological Changes as Those Seen in Human Malaria. Am J Trop Med Hyg 2017; 97:548-555. [PMID: 28829738 DOI: 10.4269/ajtmh.16-0742] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Histopathological data collected from patients with severe malaria have been instrumental for studying malaria pathogenesis. Animal models of malaria are critical to complement such studies. Here, the histopathological changes observed in a rhesus macaque with severe and complicated Plasmodium cynomolgi malaria are reported. The animal presented with thrombocytopenia, severe anemia, and hyperparasitemia during the acute infection. The macaque was given subcurative antimalarial treatment, fluid support, and a blood transfusion to treat the clinical complications, but at the time of transfusion, kidney function was compromised. These interventions did not restore kidney function, and the animal was euthanized due to irreversible renal failure. Gross pathological and histological examinations revealed that the lungs, kidneys, liver, spleen, and bone marrow exhibited abnormalities similar to those described in patients with malaria. Overall, this case report illustrates the similarities in the pathophysiological complications that can occur in human malaria and cynomolgi malaria in rhesus macaques.
Collapse
Affiliation(s)
- Chester J Joyner
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - The MaHPIC Consortium
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Jennifer S Wood
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Alberto Moreno
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, Georgia.,Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia.,Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Anapatricia Garcia
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Mary R Galinski
- Malaria Host-Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia.,Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia.,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
36
|
Anderson DC, Lapp SA, Barnwell JW, Galinski MR. A large scale Plasmodium vivax- Saimiri boliviensis trophozoite-schizont transition proteome. PLoS One 2017; 12:e0182561. [PMID: 28829774 PMCID: PMC5567661 DOI: 10.1371/journal.pone.0182561] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 07/20/2017] [Indexed: 11/18/2022] Open
Abstract
Plasmodium vivax is a complex protozoan parasite with over 6,500 genes and stage-specific differential expression. Much of the unique biology of this pathogen remains unknown, including how it modifies and restructures the host reticulocyte. Using a recently published P. vivax reference genome, we report the proteome from two biological replicates of infected Saimiri boliviensis host reticulocytes undergoing transition from the late trophozoite to early schizont stages. Using five database search engines, we identified a total of 2000 P. vivax and 3487 S. boliviensis proteins, making this the most comprehensive P. vivax proteome to date. PlasmoDB GO-term enrichment analysis of proteins identified at least twice by a search engine highlighted core metabolic processes and molecular functions such as glycolysis, translation and protein folding, cell components such as ribosomes, proteasomes and the Golgi apparatus, and a number of vesicle and trafficking related clusters. Database for Annotation, Visualization and Integrated Discovery (DAVID) v6.8 enriched functional annotation clusters of S. boliviensis proteins highlighted vesicle and trafficking-related clusters, elements of the cytoskeleton, oxidative processes and response to oxidative stress, macromolecular complexes such as the proteasome and ribosome, metabolism, translation, and cell death. Host and parasite proteins potentially involved in cell adhesion were also identified. Over 25% of the P. vivax proteins have no functional annotation; this group includes 45 VIR members of the large PIR family. A number of host and pathogen proteins contained highly oxidized or nitrated residues, extending prior trophozoite-enriched stage observations from S. boliviensis infections, and supporting the possibility of oxidative stress in relation to the disease. This proteome significantly expands the size and complexity of the known P. vivax and Saimiri host iRBC proteomes, and provides in-depth data that will be valuable for ongoing research on this parasite’s biology and pathogenesis.
Collapse
Affiliation(s)
- D. C. Anderson
- Bioscience Division, SRI International, Harrisonburg, VA, United States of America
- * E-mail:
| | - Stacey A. Lapp
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - John W. Barnwell
- Malaria Branch, Division of Parasitic Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| | - Mary R. Galinski
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States of America
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States of America
| |
Collapse
|
37
|
Strict tropism for CD71 +/CD234 + human reticulocytes limits the zoonotic potential of Plasmodium cynomolgi. Blood 2017; 130:1357-1363. [PMID: 28698207 DOI: 10.1182/blood-2017-02-764787] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/30/2017] [Indexed: 01/25/2023] Open
Abstract
Two malaria parasites of Southeast Asian macaques, Plasmodium knowlesi and P cynomolgi, can infect humans experimentally. In Malaysia, where both species are common, zoonotic knowlesi malaria has recently become dominant, and cases are recorded throughout the region. By contrast, to date, only a single case of naturally acquired P cynomolgi has been found in humans. In this study, we show that whereas P cynomolgi merozoites invade monkey red blood cells indiscriminately in vitro, in humans, they are restricted to reticulocytes expressing both transferrin receptor 1 (Trf1 or CD71) and the Duffy antigen/chemokine receptor (DARC or CD234). This likely contributes to the paucity of detectable zoonotic cynomolgi malaria. We further describe postinvasion morphologic and rheologic alterations in P cynomolgi-infected human reticulocytes that are strikingly similar to those observed for P vivax These observations stress the value of P cynomolgi as a model in the development of blood stage vaccines against vivax malaria.
Collapse
|
38
|
The unhealthy attraction of Plasmodium vivax to reticulocytes expressing transferrin receptor 1 (CD71). Int J Parasitol 2017; 47:379-383. [PMID: 28414070 DOI: 10.1016/j.ijpara.2017.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 11/24/2022]
Abstract
The majority of malaria parasite species prefer to invade reticulocytes, the most infamous being Plasmodium vivax. While the absence of an in vitro continuous culture method has hampered the study of P. vivax invasion biology, studies utilising primate models and ex vivo assays have provided some important insights. Most importantly, P. vivax merozoites have a strong preference for a subset of immature erythrocytes characterised by the expression of the transferrin receptor (CD71). This current opinion piece on P. vivax merozoite invasion highlights important gaps in our understanding of how this parasite recognises and enters reticulocytes, and discusses some recent conceptual advances in P. vivax invasion biology.
Collapse
|