1
|
Wu J, Oguz C, Teklemichael AA, Xu F, Stadler RV, Lucky AB, Liu S, Kaneko O, Lack J, Su XZ. Comparative genomics of Plasmodium yoelii nigeriensis N67 and N67C: genome-wide polymorphisms, differential gene expression, and drug resistance. BMC Genomics 2024; 25:1035. [PMID: 39497038 PMCID: PMC11536827 DOI: 10.1186/s12864-024-10961-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/25/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND The study of rodent malaria parasites has significantly advanced our understanding of malaria parasite biology and host responses to parasite infections. There are four well-characterized rodent malaria parasite species (Plasmodium yoelii, P. chabaudi, P. berghei, and P. vinckei). Each species also has multiple strains that cause different disease phenotypes. P. yoelii nigeriensis N67C and N67, two isogenic parasites, are particularly intriguing as they differ in virulence and incite different immune responses in mice. The genome of the N67 parasite has been assembled recently, but not that of N67C. This study used PacBio HiFi sequencing data to assemble the N67C genome, compared the two genomes, and performed RNA sequencing to identify polymorphisms and differentially expressed genes (DEGs). RESULTS The assembled N67C parasite genome consisted of 16 scaffolds and three contigs of approximately 22.5 Mb with 100% and 96.6% completeness based on well-characterized single-copy orthologs specific to the Apicomplexa phylum and the Plasmodium genus, respectively. A comparison between the annotated N67C and N67 genomes revealed 133 single nucleotide polymorphisms (SNPs) and 75 indels. Among the polymorphic sites, an S (N67) to N (N67C) amino acid substitution at position 114 (S114N) in the dihydrofolate reductase-thymidylate synthase (DHFR-TS) confers resistance to pyrimethamine in mice. Additionally, 60 differentially expressed single-copy genes (DEGs) were detected after comparing mRNA levels between the two parasites. Starting with the predicted and annotated 5,681 N67C and 5,749 N67 genes, we identified 4,641 orthogroups that included at least one gene from the four P. yoelii parasites (N67, N67C, 17X, and YM), whereas 758 orthogroups showed subspecies or strain-specific patterns. CONCLUSION The identification of polymorphic sites between the N67 and N67C genomes, along with the detection of the DEGs, may provide crucial insights into the variations in parasite drug responses and disease severity between these two isogenic parasites. The functional characterization of these genetic differences and candidate genes will deepen our understanding of disease mechanisms and pave the way for developing more effective control measures against malaria.
Collapse
Affiliation(s)
- Jian Wu
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, 20852, USA
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Awet Alem Teklemichael
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, 20852, USA
| | - Fangzheng Xu
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, 20852, USA
| | - Rachel V Stadler
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, 20852, USA
| | - Amuza Byaruhanga Lucky
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523, Japan
| | - Shengfa Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523, Japan
| | - Justin Lack
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xin-Zhuan Su
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, 20852, USA.
| |
Collapse
|
2
|
Yadav N, Kalata AC, Reynolds RA, Raappana A, Sather DN, Murphy SC. Identifying Plasmodium P36 and P52 antigens for co-administration with circumsporozoite protein to enhance vaccine efficacy. RESEARCH SQUARE 2024:rs.3.rs-4909396. [PMID: 39399676 PMCID: PMC11469399 DOI: 10.21203/rs.3.rs-4909396/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Vaccines targeting the complex pre-erythrocytic stage of Plasmodium parasites may benefit from inclusion of multiple antigens. However, discerning protective effects can be difficult because newer candidates may not be as protective as leading antigens like the circumsporozoite protein (CSP) in the conventional pre-clinical mouse model. We developed a modified mouse model challenge strategy that maximizes the contribution of T cells induced by novel candidate antigens at the sporozoite challenge time point and used this approach to test Plasmodium P36 and P52 vaccine candidates alone and in concert with non-protective doses of CSP. Co-administration of P36 and/or P52 with CSP achieved 80-100% sterile protection in mice, compared to only 7-30% protection for each individual antigen. P36 and P52 vaccination induced murine CD4+ and CD8+ T cell responses, but not antibody responses. This study adds P36 and P52 as promising vaccine antigens that may enhance protection achieved by CSP vaccination.
Collapse
Affiliation(s)
- Naveen Yadav
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
| | - Anya C. Kalata
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
| | - Rebekah A. Reynolds
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
| | - Andrew Raappana
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - D. Noah Sather
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| |
Collapse
|
3
|
Hazzard B, Sá JM, Bogale HN, Pascini TV, Ellis AC, Amin S, Armistead JS, Adams JH, Wellems TE, Serre D. Single-cell analyses of polyclonal Plasmodium vivax infections and their consequences on parasite transmission. Nat Commun 2024; 15:7625. [PMID: 39223117 PMCID: PMC11369214 DOI: 10.1038/s41467-024-51949-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Most Plasmodium vivax infections contain genetically distinct parasites, but the consequences of this polyclonality on the development of asexual parasites, their sexual differentiation, and their transmission remain unknown. We describe infections of Saimiri monkeys with two strains of P. vivax and the analyses of 80,024 parasites characterized by single cell RNA sequencing and individually genotyped. In our model, consecutive inoculations fail to establish polyclonal infections. By contrast, simultaneous inoculations of two strains lead to sustained polyclonal infections, although without detectable differences in parasite regulation or sexual commitment. Analyses of sporozoites dissected from mosquitoes fed on coinfected monkeys show that all genotypes are successfully transmitted to mosquitoes. However, after sporozoite inoculation, not all genotypes contribute to the subsequent blood infections, highlighting an important bottleneck during pre-erythrocytic development. Overall, these studies provide new insights on the mechanisms regulating the establishment of polyclonal P. vivax infections and their consequences for disease transmission.
Collapse
Affiliation(s)
- Brittany Hazzard
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Juliana M Sá
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Haikel N Bogale
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tales V Pascini
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Angela C Ellis
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shuchi Amin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer S Armistead
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
- Center for Global Health and Inter-Disciplinary Research, College of Public Health, University of South Florida, Tampa, USA
| | - John H Adams
- Center for Global Health and Inter-Disciplinary Research, College of Public Health, University of South Florida, Tampa, USA
| | - Thomas E Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Serre
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Hazzard B, Sá JM, Bogale HN, Pascini TV, Ellis AC, Amin S, Armistead JS, Adams JH, Wellems TE, Serre D. Single-cell analyses of polyclonal Plasmodium vivax infections and their consequences on parasite transmission. RESEARCH SQUARE 2024:rs.3.rs-3888175. [PMID: 38410426 PMCID: PMC10896380 DOI: 10.21203/rs.3.rs-3888175/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Most Plasmodium vivax infections contain genetically distinct parasites, but the consequences of this polyclonality on the development of asexual parasites, their sexual differentiation, and their transmission remain unknown. We describe infections of Saimiri monkeys with two strains of P. vivax and the analyses of 117,350 parasites characterized by single cell RNA sequencing and individually genotyped. In our model, consecutive inoculations fail to establish polyclonal infections. By contrast, simultaneous inoculations of two strains lead to sustained polyclonal infections, although without detectable differences in parasite regulation or sexual commitment. Analyses of sporozoites dissected from mosquitoes fed on coinfected monkeys show that all genotypes are successfully transmitted to mosquitoes. However, after sporozoite inoculation, not all genotypes contribute to the subsequent blood infections, highlighting an important bottleneck during pre-erythrocytic development. Overall, these studies provide new insights on the mechanisms regulating the establishment of polyclonal P. vivax infections and their consequences for disease transmission.
Collapse
Affiliation(s)
- Brittany Hazzard
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Juliana M. Sá
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Haikel N. Bogale
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tales V. Pascini
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Angela C. Ellis
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Shuchi Amin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jennifer S. Armistead
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Center for Global Health and Inter-Disciplinary Research, College of Public Health, University of South Florida, Tampa, USA
| | - John H. Adams
- Center for Global Health and Inter-Disciplinary Research, College of Public Health, University of South Florida, Tampa, USA
| | - Thomas E. Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - David Serre
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Lead contact
| |
Collapse
|
5
|
Locke E, Flores-Garcia Y, Mayer BT, MacGill RS, Borate B, Salgado-Jimenez B, Gerber MW, Mathis-Torres S, Shapiro S, King CR, Zavala F. Establishing RTS,S/AS01 as a benchmark for comparison to next-generation malaria vaccines in a mouse model. NPJ Vaccines 2024; 9:29. [PMID: 38341502 DOI: 10.1038/s41541-024-00819-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
New strategies are needed to reduce the incidence of malaria, and promising approaches include vaccines targeting the circumsporozoite protein (CSP). To improve upon the malaria vaccine, RTS,S/AS01, it is essential to standardize preclinical assays to measure the potency of next-generation vaccines against this benchmark. We focus on RTS,S/AS01-induced antibody responses and functional activity in conjunction with robust statistical analyses. Transgenic Plasmodium berghei sporozoites containing full-length P. falciparum CSP (tgPb-PfCSP) allow two assessments of efficacy: quantitative reduction in liver infection following intravenous challenge, and sterile protection from mosquito bite challenge. Two or three doses of RTS,S/AS01 were given intramuscularly at 3-week intervals, with challenge 2-weeks after the last vaccination. Minimal inter- and intra-assay variability indicates the reproducibility of the methods. Importantly, the range of this model is suitable for screening more potent vaccines. Levels of induced anti-CSP antibody 2A10 equivalency were also associated with activity: 105 μg/mL (95% CI: 68.8, 141) reduced liver infection by 50%, whereas 285 μg/mL (95% CI: 166, 404) is required for 50% sterile protection from mosquito bite challenge. Additionally, the liver burden model was able to differentiate between protected and non-protected human plasma samples from a controlled human malaria infection study, supporting these models' relevance and predictive capability. Comparison in animal models of CSP-based vaccine candidates to RTS,S/AS01 is now possible under well controlled conditions. Assessment of the quality of induced antibodies, likely a determinant of durability of protection in humans, should be possible using these methods.
Collapse
Affiliation(s)
- Emily Locke
- Center for Vaccine Innovation and Access, PATH, Washington, DC, 20001, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Bryan T Mayer
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Randall S MacGill
- Center for Vaccine Innovation and Access, PATH, Washington, DC, 20001, USA
| | - Bhavesh Borate
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Berenice Salgado-Jimenez
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Monica W Gerber
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Shamika Mathis-Torres
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sarah Shapiro
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - C Richter King
- Center for Vaccine Innovation and Access, PATH, Washington, DC, 20001, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
6
|
Schäfer TM, Pessanha de Carvalho L, Inoue J, Kreidenweiss A, Held J. The problem of antimalarial resistance and its implications for drug discovery. Expert Opin Drug Discov 2024; 19:209-224. [PMID: 38108082 DOI: 10.1080/17460441.2023.2284820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Malaria remains a devastating infectious disease with hundreds of thousands of casualties each year. Antimalarial drug resistance has been a threat to malaria control and elimination for many decades and is still of concern today. Despite the continued effectiveness of current first-line treatments, namely artemisinin-based combination therapies, the emergence of drug-resistant parasites in Southeast Asia and even more alarmingly the occurrence of resistance mutations in Africa is of great concern and requires immediate attention. AREAS COVERED A comprehensive overview of the mechanisms underlying the acquisition of drug resistance in Plasmodium falciparum is given. Understanding these processes provides valuable insights that can be harnessed for the development and selection of novel antimalarials with reduced resistance potential. Additionally, strategies to mitigate resistance to antimalarial compounds on the short term by using approved drugs are discussed. EXPERT OPINION While employing strategies that utilize already approved drugs may offer a prompt and cost-effective approach to counter antimalarial drug resistance, it is crucial to recognize that only continuous efforts into the development of novel antimalarial drugs can ensure the successful treatment of malaria in the future. Incorporating resistance propensity assessment during this developmental process will increase the likelihood of effective and enduring malaria treatments.
Collapse
Affiliation(s)
| | | | - Juliana Inoue
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Jana Held
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| |
Collapse
|
7
|
Du EJ, Muench MO. A Monocytic Barrier to the Humanization of Immunodeficient Mice. Curr Stem Cell Res Ther 2024; 19:959-980. [PMID: 37859310 PMCID: PMC10997744 DOI: 10.2174/011574888x263597231001164351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023]
Abstract
Mice with severe immunodeficiencies have become very important tools for studying foreign cells in an in vivo environment. Xenotransplants can be used to model cells from many species, although most often, mice are humanized through the transplantation of human cells or tissues to meet the needs of medical research. The development of immunodeficient mice is reviewed leading up to the current state-of-the-art strains, such as the NOD-scid-gamma (NSG) mouse. NSG mice are excellent hosts for human hematopoietic stem cell transplants or immune reconstitution through transfusion of human peripheral blood mononuclear cells. However, barriers to full hematopoietic engraftment still remain; notably, the survival of human cells in the circulation is brief, which limits overall hematological and immune reconstitution. Reports have indicated a critical role for monocytic cells - monocytes, macrophages, and dendritic cells - in the clearance of xenogeneic cells from circulation. Various aspects of the NOD genetic background that affect monocytic cell growth, maturation, and function that are favorable to human cell transplantation are discussed. Important receptors, such as SIRPα, that form a part of the innate immune system and enable the recognition and phagocytosis of foreign cells by monocytic cells are reviewed. The development of humanized mouse models has taken decades of work in creating more immunodeficient mice, genetic modification of these mice to express human genes, and refinement of transplant techniques to optimize engraftment. Future advances may focus on the monocytic cells of the host to find ways for further engraftment and survival of xenogeneic cells.
Collapse
Affiliation(s)
- Emily J. Du
- Vitalant Research Institute, 360 Spear Street, Suite 200, San Francisco, CA, 94105, USA
| | - Marcus O. Muench
- Vitalant Research Institute, 360 Spear Street, Suite 200, San Francisco, CA, 94105, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
8
|
Nicholas J, Kolli SK, Subramani PA, De SL, Ogbondah MM, Barnes SJ, Ntumngia FB, Adams JH. Comparative analyses of functional antibody-mediated inhibition with anti-circumsporozoite monoclonal antibodies against transgenic Plasmodium berghei. Malar J 2023; 22:335. [PMID: 37936181 PMCID: PMC10629016 DOI: 10.1186/s12936-023-04765-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Acquired functional inhibitory antibodies are one of several humoral immune mechanisms used to neutralize foreign pathogens. In vitro bioassays are useful tools for quantifying antibody-mediated inhibition and evaluating anti-parasite immune antibodies. However, a gap remains in understanding of how antibody-mediated inhibition in vitro translates to inhibition in vivo. In this study, two well-characterized transgenic Plasmodium berghei parasite lines, PbmCh-luc and Pb-PfCSP(r), and murine monoclonal antibodies (mAbs) specific to P. berghei and Plasmodium falciparum circumsporozoite protein (CSP), 3D11 and 2A10, respectively, were used to evaluate antibody-mediated inhibition of parasite development in both in vitro and in vivo functional assays. METHODS IC50 values of mAbs were determined using an established inhibition of liver-stage development assay (ILSDA). For the in vivo inhibition assay, mice were passively immunized by transfer of the mAbs and subsequently challenged with 5.0 × 103 sporozoites via tail vein injection. The infection burden in both assays was quantified by luminescence and qRT-PCR of P. berghei 18S rRNA normalized to host GAPDH. RESULTS The IC50 values quantified by relative luminescence of mAbs 3D11 and 2A10 were 0.396 µg/ml and 0.093 µg/ml, respectively, against transgenic lines in vitro. Using the highest (> 90%) inhibitory antibody concentrations in a passive transfer, an IC50 of 233.8 µg/ml and 181.5 µg/ml for mAbs 3D11 and 2A10, respectively, was observed in vivo. At 25 µg (250 µg/ml), the 2A10 antibody significantly inhibited liver burden in mice compared to control. Additionally, qRT-PCR of P. berghei 18S rRNA served as a secondary validation of liver burden quantification. CONCLUSIONS Results from both experimental models, ILSDA and in vivo challenge, demonstrated that increased concentrations of the homologous anti-CSP repeat mAbs increased parasite inhibition. However, differences in antibody IC50 values between parasite lines did not allow a direct correlation between the inhibition of sporozoite invasion in vitro by ILSDA and the inhibition of mouse liver stage burden. Further studies are needed to establish the conditions for confident predictions for the in vitro ILSDA to be a predictor of in vivo outcomes using this model system.
Collapse
Affiliation(s)
- Justin Nicholas
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Surendra Kumar Kolli
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Pradeep Annamalai Subramani
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Sai Lata De
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
- Department of Infectious Disease & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Madison M Ogbondah
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Samantha J Barnes
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Francis Babila Ntumngia
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - John H Adams
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
9
|
Basco LK. Cultivation of Asexual Intraerythrocytic Stages of Plasmodium falciparum. Pathogens 2023; 12:900. [PMID: 37513747 PMCID: PMC10384318 DOI: 10.3390/pathogens12070900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Successfully developed in 1976, the continuous in vitro culture of Plasmodium falciparum has many applications in the field of malaria research. It has become an important experimental model that directly uses a human pathogen responsible for a high prevalence of morbidity and mortality in many parts of the world and is a major source of biological material for immunological, biochemical, molecular, and pharmacological studies. Until present, the basic techniques described by Trager and Jensen and Haynes et al. remain unchanged in many malaria research laboratories. Nonetheless, different factors, including culture media, buffers, serum substitutes and supplements, sources of erythrocytes, and conditions of incubation (especially oxygen concentration), have been modified by different investigators to adapt the original technique in their laboratories or enhance the in vitro growth of the parasites. The possible effects and benefits of these modifications for the continuous cultivation of asexual intraerythrocytic stages of P. falciparum, as well as future challenges in developing a serum-free cultivation system and axenic cultures, are discussed.
Collapse
Affiliation(s)
- Leonardo K Basco
- Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique-Hôpitaux de Marseille (AP-HM), Service de Santé des Armées (SSA), Unité Mixte de Recherche (UMR) Vecteurs-Infections Tropicales et Méditerranéennes (VITROME), 13005 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
| |
Collapse
|
10
|
Marques-da-Silva C, Poudel B, Baptista RP, Peissig K, Hancox LS, Shiau JC, Pewe LL, Shears MJ, Kanneganti TD, Sinnis P, Kyle DE, Gurung P, Harty JT, Kurup SP. AIM2 sensors mediate immunity to Plasmodium infection in hepatocytes. Proc Natl Acad Sci U S A 2023; 120:e2210181120. [PMID: 36595704 PMCID: PMC9926219 DOI: 10.1073/pnas.2210181120] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/18/2022] [Indexed: 01/05/2023] Open
Abstract
Malaria, caused by Plasmodium parasites is a severe disease affecting millions of people around the world. Plasmodium undergoes obligatory development and replication in the hepatocytes, before initiating the life-threatening blood-stage of malaria. Although the natural immune responses impeding Plasmodium infection and development in the liver are key to controlling clinical malaria and transmission, those remain relatively unknown. Here we demonstrate that the DNA of Plasmodium parasites is sensed by cytosolic AIM2 (absent in melanoma 2) receptors in the infected hepatocytes, resulting in Caspase-1 activation. Remarkably, Caspase-1 was observed to undergo unconventional proteolytic processing in hepatocytes, resulting in the activation of the membrane pore-forming protein, Gasdermin D, but not inflammasome-associated proinflammatory cytokines. Nevertheless, this resulted in the elimination of Plasmodium-infected hepatocytes and the control of malaria infection in the liver. Our study uncovers a pathway of natural immunity critical for the control of malaria in the liver.
Collapse
Affiliation(s)
- Camila Marques-da-Silva
- Department of Cellular Biology, University of Georgia, Athens, GA30605
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA30605
| | - Barun Poudel
- Department of Internal Medicine, University of Iowa, Iowa City, IA52242
| | - Rodrigo P. Baptista
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA30605
- Institute of Bioinformatics, University of Georgia, Athens, GA30605
| | - Kristen Peissig
- Department of Cellular Biology, University of Georgia, Athens, GA30605
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA30605
| | - Lisa S. Hancox
- Department of Pathology, University of Iowa, Iowa City, IA52242
| | - Justine C. Shiau
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA30605
- Department of Infectious Diseases, University of Georgia, Athens, GA30605
| | - Lecia L. Pewe
- Department of Pathology, University of Iowa, Iowa City, IA52242
| | - Melanie J. Shears
- Johns Hopkins Malaria Research Institute, Johns Hopkins University, Baltimore, MD21205
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD21205
| | | | - Photini Sinnis
- Johns Hopkins Malaria Research Institute, Johns Hopkins University, Baltimore, MD21205
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD21205
| | - Dennis E. Kyle
- Department of Cellular Biology, University of Georgia, Athens, GA30605
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA30605
- Department of Infectious Diseases, University of Georgia, Athens, GA30605
| | - Prajwal Gurung
- Department of Internal Medicine, University of Iowa, Iowa City, IA52242
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA52242
| | - John T. Harty
- Department of Pathology, University of Iowa, Iowa City, IA52242
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA52242
| | - Samarchith P. Kurup
- Department of Cellular Biology, University of Georgia, Athens, GA30605
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA30605
| |
Collapse
|
11
|
Tucker MS, Khan A, Jenkins MC, Dubey JP, Rosenthal BM. Hastening Progress in Cyclospora Requires Studying Eimeria Surrogates. Microorganisms 2022; 10:1977. [PMID: 36296256 PMCID: PMC9608778 DOI: 10.3390/microorganisms10101977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Cyclospora cayetanensis is an enigmatic human parasite that sickens thousands of people worldwide. The scarcity of research material and lack of any animal model or cell culture system slows research, denying the produce industry, epidemiologists, and regulatory agencies of tools that might aid diagnosis, risk assessment, and risk abatement. Fortunately, related species offer a strong foundation when used as surrogates to study parasites of this type. Species of Eimeria lend themselves especially well as surrogates for C. cayetanensis. Those Eimeria that infect poultry can be produced in abundance, share many biological features with Cyclospora, pose no risk to the health of researchers, and can be studied in their natural hosts. Here, we overview the actual and potential uses of such surrogates to advance understanding of C. cayetanensis biology, diagnostics, control, and genomics, focusing on opportunities to improve prevention, surveillance, risk assessment, and risk reduction. Studying Eimeria surrogates accelerates progress, closing important research gaps and refining promising tools for producers and food safety regulators to monitor and ameliorate the food safety risks imposed by this emerging, enigmatic parasite.
Collapse
Affiliation(s)
| | | | | | | | - Benjamin M. Rosenthal
- Animal Parasitic Disease Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, BARC-East, Beltsville, MD 20705, USA
| |
Collapse
|
12
|
Das A, Sahu W, Ojha DK, Reddy KS, Suar M. Comparative Analysis of Host Metabolic Alterations in Murine Malaria Models with Uncomplicated or Severe Malaria. J Proteome Res 2022; 21:2261-2276. [PMID: 36169658 DOI: 10.1021/acs.jproteome.2c00123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Malaria varies in severity, with complications ranging from uncomplicated to severe malaria. Severe malaria could be attributed to peripheral hyperparasitemia or cerebral malaria. The metabolic interactions between the host and Plasmodium species are yet to be understood during these infections of varied pathology and severity. An untargeted metabolomics approach utilizing the liquid chromatography-mass spectrometry platform has been used to identify the affected host metabolic pathways and associated metabolites in the serum of murine malaria models with uncomplicated malaria, hyperparasitemia, and experimental cerebral malaria. We report that mice with malaria share similar metabolic attributes like higher levels of bile acids, bile pigments, and steroid hormones that have been reported for human malaria infections. Moreover, in severe malaria, upregulated levels of metabolites like phenylalanine, histidine, valine, pipecolate, ornithine, and pantothenate, with decreased levels of arginine and hippurate, were observed. Metabolites of sphingolipid metabolism were upregulated in experimental cerebral malaria. Higher levels of 20-hydroxy-leukotriene B4 and epoxyoctadecamonoenoic acids were found in uncomplicated malaria, with lower levels observed for experimental cerebral malaria. Our study provides insights into host biology during different pathological stages of malaria disease and would be useful for the selection of animal models for evaluating diagnostic and therapeutic interventions against malaria. The raw data files are available via MetaboLights with the identifier MTBLS4387.
Collapse
Affiliation(s)
- Aleena Das
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India.,Technology Business Incubator, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India
| | - Welka Sahu
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India
| | - Deepak Kumar Ojha
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India
| | - K Sony Reddy
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India
| | - Mrutyunjay Suar
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India.,Technology Business Incubator, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India
| |
Collapse
|
13
|
Shaw CL, Kennedy DA. Developing an empirical model for spillover and emergence: Orsay virus host range in Caenorhabditis. Proc Biol Sci 2022; 289:20221165. [PMID: 36126684 PMCID: PMC9489279 DOI: 10.1098/rspb.2022.1165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
A lack of tractable experimental systems in which to test hypotheses about the ecological and evolutionary drivers of disease spillover and emergence has limited our understanding of these processes. Here we introduce a promising system: Caenorhabditis hosts and Orsay virus, a positive-sense single-stranded RNA virus that naturally infects C. elegans. We assayed species across the Caenorhabditis tree and found Orsay virus susceptibility in 21 of 84 wild strains belonging to 14 of 44 species. Confirming patterns documented in other systems, we detected effects of host phylogeny on susceptibility. We then tested whether susceptible strains were capable of transmitting Orsay virus by transplanting exposed hosts and determining whether they transmitted infection to conspecifics during serial passage. We found no evidence of transmission in 10 strains (virus undetectable after passaging in all replicates), evidence of low-level transmission in 5 strains (virus lost between passage 1 and 5 in at least one replicate) and evidence of sustained transmission in 6 strains (including all three experimental C. elegans strains) in at least one replicate. Transmission was strongly associated with viral amplification in exposed populations. Variation in Orsay virus susceptibility and transmission among Caenorhabditis strains suggests that the system could be powerful for studying spillover and emergence.
Collapse
Affiliation(s)
- Clara L. Shaw
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - David A. Kennedy
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
14
|
Kobia FM, Maiti K, Obimbo MM, Smith R, Gitaka J. Potential pharmacologic interventions targeting TLR signaling in placental malaria. Trends Parasitol 2022; 38:513-524. [DOI: 10.1016/j.pt.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
|
15
|
Carey MA, Medlock GL, Stolarczyk M, Petri WA, Guler JL, Papin JA. Comparative analyses of parasites with a comprehensive database of genome-scale metabolic models. PLoS Comput Biol 2022; 18:e1009870. [PMID: 35196325 PMCID: PMC8901074 DOI: 10.1371/journal.pcbi.1009870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/07/2022] [Accepted: 01/27/2022] [Indexed: 01/01/2023] Open
Abstract
Protozoan parasites cause diverse diseases with large global impacts. Research on the pathogenesis and biology of these organisms is limited by economic and experimental constraints. Accordingly, studies of one parasite are frequently extrapolated to infer knowledge about another parasite, across and within genera. Model in vitro or in vivo systems are frequently used to enhance experimental manipulability, but these systems generally use species related to, yet distinct from, the clinically relevant causal pathogen. Characterization of functional differences among parasite species is confined to post hoc or single target studies, limiting the utility of this extrapolation approach. To address this challenge and to accelerate parasitology research broadly, we present a functional comparative analysis of 192 genomes, representing every high-quality, publicly-available protozoan parasite genome including Plasmodium, Toxoplasma, Cryptosporidium, Entamoeba, Trypanosoma, Leishmania, Giardia, and other species. We generated an automated metabolic network reconstruction pipeline optimized for eukaryotic organisms. These metabolic network reconstructions serve as biochemical knowledgebases for each parasite, enabling qualitative and quantitative comparisons of metabolic behavior across parasites. We identified putative differences in gene essentiality and pathway utilization to facilitate the comparison of experimental findings and discovered that phylogeny is not the sole predictor of metabolic similarity. This knowledgebase represents the largest collection of genome-scale metabolic models for both pathogens and eukaryotes; with this resource, we can predict species-specific functions, contextualize experimental results, and optimize selection of experimental systems for fastidious species.
Collapse
Affiliation(s)
- Maureen A. Carey
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- * E-mail: (MAC); (JP)
| | - Gregory L. Medlock
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Michał Stolarczyk
- Department of Biology, University of Virginia, Charlottesville, Virginia, United States of America
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - William A. Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Jennifer L. Guler
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jason A. Papin
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- * E-mail: (MAC); (JP)
| |
Collapse
|
16
|
Simwela NV, Waters AP. Current status of experimental models for the study of malaria. Parasitology 2022; 149:1-22. [PMID: 35357277 PMCID: PMC9378029 DOI: 10.1017/s0031182021002134] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Infection by malaria parasites (Plasmodium spp.) remains one of the leading causes of morbidity and mortality, especially in tropical regions of the world. Despite the availability of malaria control tools such as integrated vector management and effective therapeutics, these measures have been continuously undermined by the emergence of vector resistance to insecticides or parasite resistance to frontline antimalarial drugs. Whilst the recent pilot implementation of the RTS,S malaria vaccine is indeed a remarkable feat, highly effective vaccines against malaria remain elusive. The barriers to effective vaccines result from the complexity of both the malaria parasite lifecycle and the parasite as an organism itself with consequent major gaps in our understanding of their biology. Historically and due to the practical and ethical difficulties of working with human malaria infections, research into malaria parasite biology has been extensively facilitated by animal models. Animals have been used to study disease pathogenesis, host immune responses and their (dys)regulation and further disease processes such as transmission. Moreover, animal models remain at the forefront of pre-clinical evaluations of antimalarial drugs (drug efficacy, mode of action, mode of resistance) and vaccines. In this review, we discuss commonly used animal models of malaria, the parasite species used and their advantages and limitations which hinder their extrapolation to actual human disease. We also place into this context the most recent developments such as organoid technologies and humanized mice.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Andrew P. Waters
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| |
Collapse
|
17
|
Azasi Y. Assay of Static Adhesion of Plasmodium falciparum-Infected Erythrocytes to Cells, Including Inhibition of the Adhesion. Methods Mol Biol 2022; 2470:515-525. [PMID: 35881371 DOI: 10.1007/978-1-0716-2189-9_39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A feature of the virulent malaria parasite, Plasmodium falciparum, is the sequestration of infected erythrocytes (IEs) to host endothelium. The IEs sequester in the microvasculature by adhesion to host cells resulting in the obstruction of blood flow and often harmful consequences in the host. IEs bind to receptors on host cells with the P. falciparum erythrocyte membrane protein 1 (PfEMP1) that is expressed on the surface of the IEs. The study of parasite cytoadhesion is essential to decipher these ligands, including types of PfEMP1 required for cytoadhesion, the receptors the IEs bind, and how they may be related to the type of malaria disease. An assay for IE adhesion to host cells, including the inhibition of cytoadhesion is described here. The assay involves the purification of IEs with knobs and binding of the IEs to a monolayer of host cells under static conditions. Compounds including proteins, antibodies or drugs can be tested for cytoadhesion inhibitory activity in the assay.
Collapse
Affiliation(s)
- Yvonne Azasi
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina , Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
18
|
Ebel ER, Uricchio LH, Petrov DA, Egan ES. Revisiting the malaria hypothesis: accounting for polygenicity and pleiotropy. Trends Parasitol 2022; 38:290-301. [PMID: 35065882 PMCID: PMC8916997 DOI: 10.1016/j.pt.2021.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 10/19/2022]
Abstract
The malaria hypothesis predicts local, balancing selection of deleterious alleles that confer strong protection from malaria. Three protective variants, recently discovered in red cell genes, are indeed more common in African than European populations. Still, up to 89% of the heritability of severe malaria is attributed to many genome-wide loci with individually small effects. Recent analyses of hundreds of genome-wide association studies (GWAS) in humans suggest that most functional, polygenic variation is pleiotropic for multiple traits. Interestingly, GWAS alleles and red cell traits associated with small reductions in malaria risk are not enriched in African populations. We propose that other selective and neutral forces, in addition to malaria prevalence, explain the global distribution of most genetic variation impacting malaria risk.
Collapse
|
19
|
Münz C. Modification of EBV-Associated Pathologies and Immune Control by Coinfections. Front Oncol 2021; 11:756480. [PMID: 34778072 PMCID: PMC8581224 DOI: 10.3389/fonc.2021.756480] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/08/2021] [Indexed: 12/19/2022] Open
Abstract
The oncogenic Epstein–Barr virus (EBV) persistently infects more than 95% of the human adult population. Even so it can readily transform human B cells after infection in vitro, it only rarely causes tumors in patients. A substantial proportion of the 1% of all human cancers that are associated with EBV occurs during coinfections, including those with the malaria parasite Plasmodium falciparum, the human immunodeficiency virus (HIV), and the also oncogenic and closely EBV-related Kaposi sarcoma-associated herpesvirus (KSHV). In this review, I will discuss how these infections interact with EBV, modify its immune control, and shape its tumorigenesis. The underlying mechanisms reveal new aspects of EBV-associated pathologies and point toward treatment possibilities for their prevention by the human immune system.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
20
|
Abstract
Extensive research conducted on mouse-human chimeras has advanced our understanding on infectious diseases including the human-malaria parasite, Plasmodium falciparum. In vitro culture of asexual-blood stage infection of P. falciparum does not answer all questions related to parasitology, pharmacology and immunology, and complex life cycle, complicated genome, evolution of drug resistance and poor diagnosis makes it difficult to understand the patho-biology of parasite. Unavailability of effective-vaccine and issues of drug resistance advocates the use of human cell/tissues reconstituted immunodeficient-mice to P. falciparum. A number of immunodeficient-strains (TK/NOG, FRG/NOD, NOD/SCID/IL-2 receptor γ chain null, NOD severe combined immunodeficiency gamma [NSG] mouse and NOD.Rag1-/- IL2Rγ-/- [NRG; DRAG]) are used for humanization purposes. Additionally, human-hematopoietic stem cells (CD34 reconstituted-NSG [human immune system]) mice support the engraftment and repopulation of immune effecters to study systemic inflammatory diseases.
Collapse
Affiliation(s)
- Rajeev K Tyagi
- Division of Cell Biology & Immunology, Biomedical Parasitology & Nano-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh, 160036, India
| |
Collapse
|
21
|
Mouhoub E, Domenech P, Ndao M, Reed MB. The Diverse Applications of Recombinant BCG-Based Vaccines to Target Infectious Diseases Other Than Tuberculosis: An Overview. Front Microbiol 2021; 12:757858. [PMID: 34745066 PMCID: PMC8566895 DOI: 10.3389/fmicb.2021.757858] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Live attenuated Bacillus Calmette-Guérin (BCG) is the world's most widely used vaccine which is mainly administered for its protection against tuberculosis (TB), particularly in young children. However, since its initial use over 100years ago, it has also proven to offer a level of protection against various other pathogens, as a consequence of its non-specific immune enhancing effects. Thus, over the past few decades, recombinant BCG (rBCG) technology has been used as a vector to create rBCG vaccines expressing heterologous antigens that elicit immunity against a range of bacterial, viral, and parasitic diseases. Our goal with this mini-review is to provide an up-to-date survey of the various techniques, approaches, and applications of rBCG-based vaccines for targeting infectious diseases other than TB.
Collapse
Affiliation(s)
- Esma Mouhoub
- The Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada
- The Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- The McGill International TB Centre, McGill University, Montreal, QC, Canada
| | - Pilar Domenech
- The Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- The McGill International TB Centre, McGill University, Montreal, QC, Canada
| | - Momar Ndao
- The Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada
- The Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- The McGill International TB Centre, McGill University, Montreal, QC, Canada
- The Department of Medicine, McGill University, Montreal, QC, Canada
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Michael B. Reed
- The Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada
- The Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- The McGill International TB Centre, McGill University, Montreal, QC, Canada
- The Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
22
|
Patra S, Singh M, Wasnik K, Pareek D, Gupta PS, Mukherjee S, Paik P. Polymeric Nanoparticle Based Diagnosis and Nanomedicine for Treatment and Development of Vaccines for Cerebral Malaria: A Review on Recent Advancement. ACS APPLIED BIO MATERIALS 2021; 4:7342-7365. [PMID: 35006689 DOI: 10.1021/acsabm.1c00635] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cerebral malaria occurs due to Plasmodium falciparum infection, which causes 228 million infections and 450,000 deaths worldwide every year. African people are mostly affected with nearly 91% cases, of which 86% are pregnant women and infants. India and Brazil are the other two countries severely suffering from malaria endemicity. Commonly used drugs have severe side effects, and unfortunately no suitable vaccine is available in the market today. In this line, this review is focused on polymeric nanomaterials and nanocapsules that can be used for the development of effective diagnostic strategies, nanomedicines, and vaccines in the management of cerebral malaria. Further, this review will help scientists and medical professionals by updating the status on the development stages of polymeric nanoparticle based diagnostics, nanomedicines, and vaccines and strategies to eradicate cerebral malaria. In addition to this, the predominant focus of this review is antimalarial agents based on polymer nanomedicines that are currently in the preclinical and clinical trial stages, and potential developments are suggested as well. This review further will have an important social and commercial impact worldwide for the development of polymeric nanomedicines and strategies for the treatment of cerebral malaria.
Collapse
Affiliation(s)
- Sukanya Patra
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| | - Monika Singh
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| | - Kirti Wasnik
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| | - Divya Pareek
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| | - Prem Shankar Gupta
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, Texas 77030, United States
| | - Pradip Paik
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| |
Collapse
|
23
|
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: 1.3] [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
|
24
|
Adamiak N, Krawczyk KT, Locht C, Kowalewicz-Kulbat M. Archaeosomes and Gas Vesicles as Tools for Vaccine Development. Front Immunol 2021; 12:746235. [PMID: 34567012 PMCID: PMC8462270 DOI: 10.3389/fimmu.2021.746235] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/27/2021] [Indexed: 12/03/2022] Open
Abstract
Archaea are prokaryotic organisms that were classified as a new domain in 1990. Archaeal cellular components and metabolites have found various applications in the pharmaceutical industry. Some archaeal lipids can be used to produce archaeosomes, a new family of liposomes that exhibit high stability to temperatures, pH and oxidative conditions. Additionally, archaeosomes can be efficient antigen carriers and adjuvants promoting humoral and cellular immune responses. Some archaea produce gas vesicles, which are nanoparticles released by the archaea that increase the buoyancy of the cells and facilitate an upward flotation in water columns. Purified gas vesicles display a great potential for bioengineering, due to their high stability, immunostimulatory properties and uptake across cell membranes. Both archaeosomes and archaeal gas vesicles are attractive tools for the development of novel drug and vaccine carriers to control various diseases. In this review we discuss the current knowledge on production, preparation methods and potential applications of archaeosomes and gas vesicles as carriers for vaccines. We give an overview of the traditional structures of these carriers and their modifications. A comparative analysis of both vaccine delivery systems, including their advantages and limitations of their use, is provided. Gas vesicle- and archaeosome-based vaccines may be powerful next-generation tools for the prevention and treatment of a wide variety of infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Natalia Adamiak
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Krzysztof T Krawczyk
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Camille Locht
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland.,Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Magdalena Kowalewicz-Kulbat
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| |
Collapse
|
25
|
Sedegah M, Hollingdale MR, Ganeshan H, Belmonte M, Huang J, Belmonte A, Inoue S, Velasco R, Hickey B, Teneza-Mora N, Lumsden J, Reyes S, Banania JG, Reyes A, Guzman I, Richie TL, Epstein JE, Villasante E. IMRAS-Immunization with radiation-attenuated Plasmodium falciparum sporozoites by mosquito bite: Cellular immunity to sporozoites, CSP, AMA1, TRAP and CelTOS. PLoS One 2021; 16:e0256396. [PMID: 34415964 PMCID: PMC8378721 DOI: 10.1371/journal.pone.0256396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/21/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Immunization with radiation-attenuated sporozoites (RAS) by mosquito bites provides >90% sterile protection against Plasmodium falciparum malaria in humans. We conducted a clinical trial based on data from previous RAS clinical trials that suggested that 800-1200 infected bites should induce ~50% protective vaccine efficacy (VE) against controlled human malaria infection (CHMI) administered three weeks after the final immunization. Two cohorts were immunized separately. VE was 55% in Cohort 1 but 90% in Cohort 2, the cohort that received a higher first dose and a reduced (fractional) fifth dose. Immune responses were better boosted by the fractional fifth dose in Cohort 2 and suggested the importance of the fractional fifth dose for increased protection in Cohort 2 responses. Three protected subjects were later boosted and were protected suggesting that protection could be extended to at least 67 weeks. METHODS The ex vivo FluoroSpot assay was used to measure peripheral IFN-γ, IL2, and IFN-γ+IL2 responses to PfNF54 sporozoites and malaria antigens CSP, AMA1, TRAP, and CelTOS using pools of synthetic overlapping 15mer peptides spanning each antigen. RESULTS There was no correlation between IFN-γ, IL2, and IFN-γ+IL2 responses to sporozoites and protection, but fold-increases between post-4th and post-5th responses greater than 1.0 occurred mostly in protected subjects. IFN-γ and IL2 responses to TRAP, CelTOS and CSP occurred only in protected subjects. Peripheral IFN-γ, IL2, and IFN-γ+IL2 responses were short-lived and low by 27 weeks post-CHMI but were restored by boosting. CONCLUSIONS These studies highlight the importance of vaccine dose and schedule for vaccine efficacy, and suggest that CSP, TRAP, AMA1 and CelTOS may be targets of protective immunity. The correlation between fold-increases in responses and protection should be explored in other vaccine trials. TRIAL REGISTRATION ClinicalTrials.gov NCT01994525.
Collapse
Affiliation(s)
- Martha Sedegah
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Michael R. Hollingdale
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Harini Ganeshan
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Maria Belmonte
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Jun Huang
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Arnel Belmonte
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Sandra Inoue
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Rachel Velasco
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Bradley Hickey
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Nimfa Teneza-Mora
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Joanne Lumsden
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Sharina Reyes
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Jo Glenna Banania
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Anatalio Reyes
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Ivelese Guzman
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation, Bethesda, Maryland, United States of America
| | - Thomas L. Richie
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Judith E. Epstein
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Eileen Villasante
- Malaria Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| |
Collapse
|
26
|
Kukla DA, Khetani SR. Bioengineered Liver Models for Investigating Disease Pathogenesis and Regenerative Medicine. Semin Liver Dis 2021; 41:368-392. [PMID: 34139785 DOI: 10.1055/s-0041-1731016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Owing to species-specific differences in liver pathways, in vitro human liver models are utilized for elucidating mechanisms underlying disease pathogenesis, drug development, and regenerative medicine. To mitigate limitations with de-differentiated cultures, bioengineers have developed advanced techniques/platforms, including micropatterned cocultures, spheroids/organoids, bioprinting, and microfluidic devices, for perfusing cell cultures and liver slices. Such techniques improve mature functions and culture lifetime of primary and stem-cell human liver cells. Furthermore, bioengineered liver models display several features of liver diseases including infections with pathogens (e.g., malaria, hepatitis C/B viruses, Zika, dengue, yellow fever), alcoholic/nonalcoholic fatty liver disease, and cancer. Here, we discuss features of bioengineered human liver models, their uses for modeling aforementioned diseases, and how such models are being augmented/adapted for fabricating implantable human liver tissues for clinical therapy. Ultimately, continued advances in bioengineered human liver models have the potential to aid the development of novel, safe, and efficacious therapies for liver disease.
Collapse
Affiliation(s)
- David A Kukla
- Deparment of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Salman R Khetani
- Deparment of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
27
|
Yamaguchi T, Katano I, Otsuka I, Ito R, Mochizuki M, Goto M, Takahashi T. Generation of Novel Human Red Blood Cell-Bearing Humanized Mouse Models Based on C3-Deficient NOG Mice. Front Immunol 2021; 12:671648. [PMID: 34386001 PMCID: PMC8353390 DOI: 10.3389/fimmu.2021.671648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/12/2021] [Indexed: 11/19/2022] Open
Abstract
Despite recent advances in immunodeficient mouse models bearing human red blood cells (hRBCs), the elimination of circulating hRBCs by residual innate immune systems remains a significant challenge. In this study, we evaluated the role of mouse complement C3 in the elimination of circulating hRBCs by developing a novel NOG substrain harboring a truncated version of the murine C3 gene (NOG-C3ΔMG2-3). Genetic C3 deletion prolonged the survival of transfused hRBCs in the circulation. Chemical depletion and functional impairment of mouse macrophages, using clodronate liposomes (Clo-lip) or gadolinium chloride (GdCl3), respectively, further extended the survival of hRBCs in NOG-C3ΔMG2-3 mice. Low GdCl3 toxicity allowed the establishment of hRBC-bearing mice, in which hRBCs survived for more than 4 weeks with transfusion once a week. In addition, erythropoiesis of human hematopoietic stem cells (hHSCs) was possible in NOG-C3ΔMG2-3/human GM-CSF-IL-3 transgenic mice with Clo-lip treatment. These findings indicate that mouse models harboring hRBCs can be achieved using NOG-C3ΔMG2-3 mice, which could facilitate studies of human diseases associated with RBCs.
Collapse
Affiliation(s)
- Takuya Yamaguchi
- Laboratory Animal Research Department, Central Institute for Experimental Animals (CIEA), Kawasaki, Japan
| | - Ikumi Katano
- Laboratory Animal Research Department, Central Institute for Experimental Animals (CIEA), Kawasaki, Japan
| | - Iyo Otsuka
- Laboratory Animal Research Department, Central Institute for Experimental Animals (CIEA), Kawasaki, Japan
| | - Ryoji Ito
- Laboratory Animal Research Department, Central Institute for Experimental Animals (CIEA), Kawasaki, Japan
| | | | - Motohito Goto
- Animal Resource & Technical Research Center, CIEA, Kawasaki, Japan
| | - Takeshi Takahashi
- Laboratory Animal Research Department, Central Institute for Experimental Animals (CIEA), Kawasaki, Japan
| |
Collapse
|
28
|
Schäfer C, Zanghi G, Vaughan AM, Kappe SHI. Plasmodium vivax Latent Liver Stage Infection and Relapse: Biological Insights and New Experimental Tools. Annu Rev Microbiol 2021; 75:87-106. [PMID: 34196569 DOI: 10.1146/annurev-micro-032421-061155] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Plasmodium vivax is the most widespread human malaria parasite, in part because it can form latent liver stages known as hypnozoites after transmission by female anopheline mosquitoes to human hosts. These persistent stages can activate weeks, months, or even years after the primary clinical infection; replicate; and initiate relapses of blood stage infection, which causes disease and recurring transmission. Eliminating hypnozoites is a substantial obstacle for malaria treatment and eradication since the hypnozoite reservoir is undetectable and unaffected by most antimalarial drugs. Importantly, in some parts of the globe where P. vivax malaria is endemic, as many as 90% of P. vivax blood stage infections are thought to be relapses rather than primary infections, rendering the hypnozoite a major driver of P. vivax epidemiology. Here, we review the biology of the hypnozoite and recent discoveries concerning this enigmatic parasite stage. We discuss treatment and prevention challenges, novel animal models to study hypnozoites and relapse, and hypotheses related to hypnozoite formation and activation. Expected final online publication date for the Annual Review of Microbiology, Volume 75 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Carola Schäfer
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , ,
| | - Gigliola Zanghi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , ,
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , , .,Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , , .,Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA.,Deparment of Global Health, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
29
|
Clark NF, Taylor-Robinson AW. An Ecologically Framed Comparison of The Potential for Zoonotic Transmission of Non-Human and Human-Infecting Species of Malaria Parasite. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2021; 94:361-373. [PMID: 34211355 PMCID: PMC8223545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The threats, both real and perceived, surrounding the development of new and emerging infectious diseases of humans are of critical concern to public health and well-being. Among these risks is the potential for zoonotic transmission to humans of species of the malaria parasite, Plasmodium, that have been considered historically to infect exclusively non-human hosts. Recently observed shifts in the mode, transmission, and presentation of malaria among several species studied are evidenced by shared vectors, atypical symptoms, and novel host-seeking behavior. Collectively, these changes indicate the presence of environmental and ecological pressures that are likely to influence the dynamics of these parasite life cycles and physiological make-up. These may be further affected and amplified by such factors as increased urban development and accelerated rate of climate change. In particular, the extended host-seeking behavior of what were once considered non-human malaria species indicates the specialist niche of human malaria parasites is not a limiting factor that drives the success of blood-borne parasites. While zoonotic transmission of non-human malaria parasites is generally considered to not be possible for the vast majority of Plasmodium species, failure to consider the feasibility of its occurrence may lead to the emergence of a potentially life-threatening blood-borne disease of humans. Here, we argue that recent trends in behavior among what were hitherto considered to be non-human malaria parasites to infect humans call for a cross-disciplinary, ecologically-focused approach to understanding the complexities of the vertebrate host/mosquito vector/malaria parasite triangular relationship. This highlights a pressing need to conduct a multi-species investigation for which we recommend the construction of a database to determine ecological differences among all known Plasmodium species, vectors, and hosts. Closing this knowledge gap may help to inform alternative means of malaria prevention and control.
Collapse
Affiliation(s)
- Nicole F. Clark
- Institute for Applied Ecology, University of Canberra,
Bruce, Australia,College of Medicine and Public Health, Flinders
University, Australia
| | - Andrew W. Taylor-Robinson
- Infectious Diseases Research Group, School of Health,
Medical & Applied Sciences, Central Queensland University, Brisbane,
Australia,College of Health & Human Sciences, Charles Darwin
University, Casuarina, Australia,To whom all correspondence should be addressed:
Prof Andrew W. Taylor-Robinson, Infectious Diseases Research Group, School of
Health, Medical & Applied Sciences, Central Queensland University, 160 Ann
Street, Brisbane, QLD 4000, Australia; Tel: +61 7 3295 1185;
; ORCID iD: https://orcid.org/0000-0001-7342-8348
| |
Collapse
|
30
|
Designing antimalarials that break into cells to lock down parasites. Proc Natl Acad Sci U S A 2021; 118:2108103118. [PMID: 34108246 DOI: 10.1073/pnas.2108103118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
31
|
Dash PK, Gorantla S, Poluektova L, Hasan M, Waight E, Zhang C, Markovic M, Edagwa B, Machhi J, Olson KE, Wang X, Mosley RL, Kevadiya B, Gendelman HE. Humanized Mice for Infectious and Neurodegenerative disorders. Retrovirology 2021; 18:13. [PMID: 34090462 PMCID: PMC8179712 DOI: 10.1186/s12977-021-00557-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022] Open
Abstract
Humanized mice model human disease and as such are used commonly for research studies of infectious, degenerative and cancer disorders. Recent models also reflect hematopoiesis, natural immunity, neurobiology, and molecular pathways that influence disease pathobiology. A spectrum of immunodeficient mouse strains permit long-lived human progenitor cell engraftments. The presence of both innate and adaptive immunity enables high levels of human hematolymphoid reconstitution with cell susceptibility to a broad range of microbial infections. These mice also facilitate investigations of human pathobiology, natural disease processes and therapeutic efficacy in a broad spectrum of human disorders. However, a bridge between humans and mice requires a complete understanding of pathogen dose, co-morbidities, disease progression, environment, and genetics which can be mirrored in these mice. These must be considered for understanding of microbial susceptibility, prevention, and disease progression. With known common limitations for access to human tissues, evaluation of metabolic and physiological changes and limitations in large animal numbers, studies in mice prove important in planning human clinical trials. To these ends, this review serves to outline how humanized mice can be used in viral and pharmacologic research emphasizing both current and future studies of viral and neurodegenerative diseases. In all, humanized mouse provides cost-effective, high throughput studies of infection or degeneration in natural pathogen host cells, and the ability to test transmission and eradication of disease.
Collapse
Affiliation(s)
- Prasanta K Dash
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Larisa Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Emiko Waight
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chen Zhang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Milica Markovic
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Xinglong Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bhavesh Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
32
|
Deng Y, Münz C. Roles of Lytic Viral Replication and Co-Infections in the Oncogenesis and Immune Control of the Epstein-Barr Virus. Cancers (Basel) 2021; 13:2275. [PMID: 34068598 PMCID: PMC8126045 DOI: 10.3390/cancers13092275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022] Open
Abstract
Epstein-Barr virus (EBV) is the prototypic human tumor virus whose continuous lifelong immune control is required to prevent lymphomagenesis in the more than 90% of the human adult population that are healthy carriers of the virus. Here, we review recent evidence that this immune control has not only to target latent oncogenes, but also lytic replication of EBV. Furthermore, genetic variations identify the molecular machinery of cytotoxic lymphocytes as essential for this immune control and recent studies in mice with reconstituted human immune system components (humanized mice) have begun to provide insights into the mechanistic role of these molecules during EBV infection. Finally, EBV often does not act in isolation to cause disease. Some of EBV infection-modulating co-infections, including human immunodeficiency virus (HIV) and Kaposi sarcoma-associated herpesvirus (KSHV), have been modeled in humanized mice. These preclinical in vivo models for EBV infection, lymphomagenesis, and cell-mediated immune control do not only promise a better understanding of the biology of this human tumor virus, but also the possibility to explore vaccine candidates against it.
Collapse
Affiliation(s)
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland;
| |
Collapse
|
33
|
Arez F, Rodrigues AF, Brito C, Alves PM. Bioengineered Liver Cell Models of Hepatotropic Infections. Viruses 2021; 13:773. [PMID: 33925701 PMCID: PMC8146083 DOI: 10.3390/v13050773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatitis viruses and liver-stage malaria are within the liver infections causing higher morbidity and mortality rates worldwide. The highly restricted tropism of the major human hepatotropic pathogens-namely, the human hepatitis B and C viruses and the Plasmodium falciparum and Plasmodium vivax parasites-has hampered the development of disease models. These models are crucial for uncovering the molecular mechanisms underlying the biology of infection and governing host-pathogen interaction, as well as for fostering drug development. Bioengineered cell models better recapitulate the human liver microenvironment and extend hepatocyte viability and phenotype in vitro, when compared with conventional two-dimensional cell models. In this article, we review the bioengineering tools employed in the development of hepatic cell models for studying infection, with an emphasis on 3D cell culture strategies, and discuss how those tools contributed to the level of recapitulation attained in the different model layouts. Examples of host-pathogen interactions uncovered by engineered liver models and their usefulness in drug development are also presented. Finally, we address the current bottlenecks, trends, and prospect toward cell models' reliability, robustness, and reproducibility.
Collapse
MESH Headings
- Animals
- Bioengineering/methods
- Cell Culture Techniques
- Disease Models, Animal
- Disease Susceptibility
- Drug Discovery
- Hepatitis/drug therapy
- Hepatitis/etiology
- Hepatitis/metabolism
- Hepatitis/pathology
- Hepatitis, Viral, Human/etiology
- Hepatitis, Viral, Human/metabolism
- Hepatitis, Viral, Human/pathology
- Hepatocytes/metabolism
- Hepatocytes/parasitology
- Hepatocytes/virology
- Host-Pathogen Interactions
- Humans
- Liver/metabolism
- Liver/parasitology
- Liver/virology
- Liver Diseases, Parasitic/etiology
- Liver Diseases, Parasitic/metabolism
- Liver Diseases, Parasitic/pathology
Collapse
Affiliation(s)
- Francisca Arez
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Ana F. Rodrigues
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Av. da República, 2780-157 Oeiras, Portugal
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
34
|
Buyon LE, Elsworth B, Duraisingh MT. The molecular basis of antimalarial drug resistance in Plasmodium vivax. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2021; 16:23-37. [PMID: 33957488 PMCID: PMC8113647 DOI: 10.1016/j.ijpddr.2021.04.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/31/2021] [Accepted: 04/08/2021] [Indexed: 01/07/2023]
Abstract
Plasmodium vivax is the most geographically widespread cause of human malaria and is responsible for the majority of cases outside of the African continent. While great progress has been made towards eliminating human malaria, drug resistant parasite strains pose a threat towards continued progress. Resistance has arisen to multiple antimalarials in P. vivax, including to chloroquine, which is currently the first line therapy for P. vivax in most regions. Despite its importance, an understanding of the molecular mechanisms of drug resistance in this species remains elusive, in large part due to the complex biology of P. vivax and the lack of in vitro culture. In this review, we will cover the extent and challenges of measuring clinical and in vitro drug resistance in P. vivax. We will consider the roles of candidate drug resistance genes. We will highlight the development of molecular approaches for studying P. vivax biology that provide the opportunity to validate the role of putative drug resistance mutations as well as identify novel mechanisms of drug resistance in this understudied parasite. Validated molecular determinants and markers of drug resistance are essential for the rapid and cost-effective monitoring of drug resistance in P. vivax, and will be useful for optimizing drug regimens and for informing drug policy in control and elimination settings. Drug resistance is emerging in Plasmodium vivax, an important cause of malaria. The complex biology of P. vivax and the limited range of research tools make it difficult to identify drug resistance. The molecular mechanisms of drug resistance in P. vivax remain elusive. This review highlights the extent of drug resistance, the putative mechanisms of resistance and new technologies for the study of P. vivax drug resistance.
Collapse
Affiliation(s)
- Lucas E Buyon
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Brendan Elsworth
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Manoj T Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, 02115, MA, USA.
| |
Collapse
|
35
|
Hopp CS, Kanatani S, Archer NK, Miller RJ, Liu H, Chiou KK, Miller LS, Sinnis P. Comparative intravital imaging of human and rodent malaria sporozoites reveals the skin is not a species-specific barrier. EMBO Mol Med 2021; 13:e11796. [PMID: 33750026 PMCID: PMC8033530 DOI: 10.15252/emmm.201911796] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 01/09/2023] Open
Abstract
Malaria infection starts with the injection of Plasmodium sporozoites into the host’s skin. Sporozoites are motile and move in the skin to find and enter blood vessels to be carried to the liver. Here, we present the first characterization of P. falciparum sporozoites in vivo, analyzing their motility in mouse skin and human skin xenografts and comparing their motility to two rodent malaria species. These data suggest that in contrast to the liver and blood stages, the skin is not a species‐specific barrier for Plasmodium. Indeed, P. falciparum sporozoites enter blood vessels in mouse skin at similar rates to the rodent malaria parasites. Furthermore, we demonstrate that antibodies targeting sporozoites significantly impact the motility of P. falciparum sporozoites in mouse skin. Though the sporozoite stage is a validated vaccine target, vaccine trials have been hampered by the lack of good animal models for human malaria parasites. Pre‐clinical screening of next‐generation vaccines would be significantly aided by the in vivo platform we describe here, expediting down‐selection of candidates prior to human vaccine trials.
Collapse
Affiliation(s)
- Christine S Hopp
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Sachie Kanatani
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevin K Chiou
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
36
|
Patel OPS, Beteck RM, Legoabe LJ. Antimalarial application of quinones: A recent update. Eur J Med Chem 2020; 210:113084. [PMID: 33333397 DOI: 10.1016/j.ejmech.2020.113084] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022]
Abstract
Atovaquone belongs to a naphthoquinone class of drugs and is used in combination with proguanil (Malarone) for the treatment of acute, uncomplicated malaria caused by Plasmodium falciparum (including chloroquine-resistant P. falciparum/P. vivax). Numerous quinone-derived compounds have attracted considerable attention in the last few decades due to their potential in antimalarial drug discovery. Several semi-synthetic derivatives of natural quinones, synthetic quinones (naphtho-/benzo-quinone, anthraquinones, thiazinoquinones), and quinone-based hybrids were explored for their in vitro and in vivo antimalarial activities. A careful literature survey revealed that this topic has not been compiled as a review article so far. Therefore, we herein summarise the recent discovery (the year 2009-2020) of quinone based antimalarial compounds in chronological order. This compilation would be very useful towards the exploration of novel quinone-derived compounds against malarial parasites with promising efficacy and lesser side effects.
Collapse
Affiliation(s)
- Om P S Patel
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa.
| | - Richard M Beteck
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Lesetja J Legoabe
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa.
| |
Collapse
|
37
|
Chatelain E, Scandale I. Animal models of Chagas disease and their translational value to drug development. Expert Opin Drug Discov 2020; 15:1381-1402. [PMID: 32812830 DOI: 10.1080/17460441.2020.1806233] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION American trypanosomiasis, better known as Chagas disease, is a global public health issue. Current treatments targeting the causative parasite, Trypanosoma cruzi, are limited to two old nitroheterocyclic compounds; new, safer drugs are needed. New tools to identify compounds suitable for parasitological cure in humans have emerged through efforts in drug discovery. AREAS COVERED Animal disease models are an integral part of the drug discovery process. There are numerous experimental models of Chagas disease described and in use; rather than going through each of these and their specific features, the authors focus on developments in recent years, in particular the imaging technologies that have dramatically changed the Chagas R&D landscape, and provide a critical view on their value and limitations for moving compounds forward into further development. EXPERT OPINION The application of new technological advances to the field of drug development for Chagas disease has led to the implementation of new and robust/standardized in vivo models that contributed to a better understanding of host/parasite interactions. These new models should also build confidence in their translational value for moving compounds forward into clinical development.
Collapse
Affiliation(s)
- Eric Chatelain
- R&D Department, Drugs for Neglected Diseases Initiative (DNDi) , Geneva, Switzerland
| | - Ivan Scandale
- R&D Department, Drugs for Neglected Diseases Initiative (DNDi) , Geneva, Switzerland
| |
Collapse
|
38
|
Raghunandan R, Mayer BT, Flores-Garcia Y, Gerber MW, Gottardo R, Jhun H, Herrera SM, Perez-Ramos DW, Locke E, King CR, Zavala F. Characterization of two in vivo challenge models to measure functional activity of monoclonal antibodies to Plasmodium falciparum circumsporozoite protein. Malar J 2020; 19:113. [PMID: 32183833 PMCID: PMC7079517 DOI: 10.1186/s12936-020-03181-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/03/2020] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND New strategies are needed to reduce the incidence of malaria, and promising approaches include the development of vaccines and monoclonal antibodies (mAbs) that target the circumsporozoite protein (CSP). To select the best candidates and speed development, it is essential to standardize preclinical assays to measure the potency of such interventions in animal models. METHODS Two assay configurations were studied using transgenic Plasmodium berghei expressing Plasmodium falciparum full-length circumsporozoite protein. The assays measured (1) reduction in parasite infection of the liver (liver burden) following an intravenous (i.v) administration of sporozoites and (2) protection from parasitaemia following mosquito bite challenge. Two human CSP mAbs, AB311 and AB317, were compared for their ability to inhibit infection. Multiple independent experiments were conducted to define assay variability and resultant impact on the ability to discriminate differences in mAb functional activity. RESULTS Overall, the assays produced highly consistent results in that all individual experiments showed greater functional activity for AB317 compared to AB311 as calculated by the dose required for 50% inhibition (ID50) as well as the serum concentration required for 50% inhibition (IC50). The data were then used to model experimental designs with adequate statistical power to rigorously screen, compare, and rank order novel anti-CSP mAbs. CONCLUSION The results indicate that in vivo assays described here can provide reliable information for comparing the functional activity of mAbs. The results also provide guidance regarding selection of the appropriate experimental design, dose selection, and group sizes.
Collapse
Affiliation(s)
- Rama Raghunandan
- PATH's Malaria Vaccine Initiative, 455 Massachusetts Avenue, NW, Suite 1000, Washington, DC, 20001, USA.
| | - Bryan T Mayer
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Monica W Gerber
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Hugo Jhun
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Vaccine Research and Development, Pfizer, Pearl River, NY, 10965, USA
| | - Sonia M Herrera
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Daniel W Perez-Ramos
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Emily Locke
- PATH's Malaria Vaccine Initiative, 455 Massachusetts Avenue, NW, Suite 1000, Washington, DC, 20001, USA
| | - C Richter King
- PATH's Malaria Vaccine Initiative, 455 Massachusetts Avenue, NW, Suite 1000, Washington, DC, 20001, USA.
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
39
|
Meehan GR, Scales HE, Osii R, De Niz M, Lawton JC, Marti M, Garside P, Craig A, Brewer JM. Developing a xenograft model of human vasculature in the mouse ear pinna. Sci Rep 2020; 10:2058. [PMID: 32029768 PMCID: PMC7004987 DOI: 10.1038/s41598-020-58650-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 01/14/2020] [Indexed: 12/31/2022] Open
Abstract
Humanised xenograft models allow for the analysis of human tissue within a physiological environment in vivo. However, current models often rely on the angiogenesis and ingrowth of recipient vasculature to perfuse tissues, preventing analysis of biological processes and diseases involving human blood vessels. This limits the effectiveness of xenografts in replicating human physiology and may lead to issues with translating findings into human research. We have designed a xenograft model of human vasculature to address this issue. Human subcutaneous fat was cultured in vitro to promote blood vessel outgrowth prior to implantation into immunocompromised mice. We demonstrate that implants survived, retained human vasculature and anastomosed with the circulatory system of the recipient mouse. Significantly, by performing transplants into the ear pinna, this system enabled intravital observation of xenografts by multiphoton microscopy, allowing us to visualise the steps leading to vascular cytoadherence of erythrocytes infected with the human parasite Plasmodium falciparum. This model represents a useful tool for imaging the interactions that occur within human tissues in vivo and permits visualization of blood flow and cellular recruitment in a system which is amenable to intervention for various studies in basic biology together with drug evaluation and mechanism of action studies.
Collapse
Affiliation(s)
- Gavin R Meehan
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Hannah E Scales
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Rowland Osii
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Mariana De Niz
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
- Instituto de Medicina Molecular, University of Lisbon, Lisbon, Portugal
| | - Jennifer C Lawton
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Matthias Marti
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Paul Garside
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Alister Craig
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - James M Brewer
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK.
| |
Collapse
|
40
|
Julien JP, Wardemann H. Antibodies against Plasmodium falciparum malaria at the molecular level. Nat Rev Immunol 2019; 19:761-775. [DOI: 10.1038/s41577-019-0209-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
|
41
|
Goh YS, McGuire D, Rénia L. Vaccination With Sporozoites: Models and Correlates of Protection. Front Immunol 2019; 10:1227. [PMID: 31231377 PMCID: PMC6560154 DOI: 10.3389/fimmu.2019.01227] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Despite continuous efforts, the century-old goal of eradicating malaria still remains. Multiple control interventions need to be in place simultaneously to achieve this goal. In addition to effective control measures, drug therapies and insecticides, vaccines are critical to reduce mortality and morbidity. Hence, there are numerous studies investigating various malaria vaccine candidates. Most of the malaria vaccine candidates are subunit vaccines. However, they have shown limited efficacy in Phase II and III studies. To date, only whole parasite formulations have been shown to induce sterile immunity in human. In this article, we review and discuss the recent developments in vaccination with sporozoites and the mechanisms of protection involved.
Collapse
Affiliation(s)
- Yun Shan Goh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore
| | - Daniel McGuire
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
42
|
Jackson S, Meeks C, Vézina A, Robey RW, Tanner K, Gottesman MM. Model systems for studying the blood-brain barrier: Applications and challenges. Biomaterials 2019; 214:119217. [PMID: 31146177 DOI: 10.1016/j.biomaterials.2019.05.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/13/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022]
Abstract
The blood-brain barrier (BBB) poses a serious impediment to the delivery of effective therapies to the central nervous system (CNS). Over time, various model systems have been crafted and used to evaluate the complexities of the BBB, which includes an impermeable physical barrier and a series of energy-dependent efflux pumps. Models of the BBB have mainly sought to assess changes in endothelial cell permeability, the role of ATP-dependent efflux transporters in drug disposition, and alterations in communication between BBB cells and the microenvironment. In the context of disease, various animal models have been utilized to examine real time BBB drug permeability, CNS dynamic changes, and overall treatment response. In this review, we outline the use of these in vitro and in vivo blood-brain barrier model systems to study normal physiology and diseased states. These current models each have their own advantages and disadvantages for studying the response of biologic processes to physiological and pathological conditions. Additional models are needed to mimic more closely the dynamic quality of the BBB, with the goal focused on potential clinical applications.
Collapse
Affiliation(s)
- Sadhana Jackson
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States.
| | - Caitlin Meeks
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Amélie Vézina
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Robert W Robey
- Multidrug Resistance Section, Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Kandice Tanner
- Tissue Morphodynamics Unit, Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Michael M Gottesman
- Multidrug Resistance Section, Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| |
Collapse
|
43
|
De Niz M, Meehan GR, Brancucci NM, Marti M, Rotureau B, Figueiredo LM, Frischknecht F. Intravital imaging of host-parasite interactions in skin and adipose tissues. Cell Microbiol 2019; 21:e13023. [PMID: 30825872 PMCID: PMC6590052 DOI: 10.1111/cmi.13023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/20/2022]
Abstract
Intravital microscopy allows the visualisation of how pathogens interact with host cells and tissues in living animals in real time. This method has enabled key advances in our understanding of host-parasite interactions under physiological conditions. A combination of genetics, microscopy techniques, and image analysis have recently facilitated the understanding of biological phenomena in living animals at cellular and subcellular resolution. In this review, we summarise findings achieved by intravital microscopy of the skin and adipose tissues upon infection with various parasites, and we present a view into possible future applications of this method.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, Heussler GroupUniversity of BernBernSwitzerland
- Wellcome Centre for Integrative ParasitologyUniversity of GlasgowGlasgowUK
| | - Gavin R. Meehan
- Wellcome Centre for Integrative ParasitologyUniversity of GlasgowGlasgowUK
| | - Nicolas M.B. Brancucci
- Malaria Gene Regulation Unit, Department of Medical Parasitology and Infection BiologySwiss Tropical and Public Health InstituteBaselSwitzerland
- University of BaselBaselSwitzerland
| | - Matthias Marti
- Wellcome Centre for Integrative ParasitologyUniversity of GlasgowGlasgowUK
| | - Brice Rotureau
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, Department of Parasites and Insect Vectors, INSERM U1201Institut PasteurParisFrance
| | - Luisa M. Figueiredo
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo AntunesUniversidade de LisboaLisbonPortugal
| | - Friedrich Frischknecht
- Integrative Parasitology, Centre for Infectious DiseasesUniversity of Heidelberg Medical SchoolHeidelbergGermany
| |
Collapse
|
44
|
De Niz M, Nacer A, Frischknecht F. Intravital microscopy: Imaging host-parasite interactions in the brain. Cell Microbiol 2019; 21:e13024. [PMID: 30830993 DOI: 10.1111/cmi.13024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/14/2019] [Accepted: 02/24/2019] [Indexed: 12/31/2022]
Abstract
Intravital fluorescence microscopy (IVM) is a powerful technique for imaging multiple organs, including the brain of living mice and rats. It enables the direct visualisation of cells in situ providing a real-life view of biological processes that in vitro systems cannot. In addition, to the technological advances in microscopy over the last decade, there have been supporting innovations in data storage and analytical packages that enable the visualisation and analysis of large data sets. Here, we review the advantages and limitations of techniques predominantly used for brain IVM, including thinned skull windows, open skull cortical windows, and a miniaturised optical system based on microendoscopic probes that can be inserted into deep tissues. Further, we explore the relevance of these techniques for the field of parasitology. Several protozoan infections are associated with neurological symptoms including Plasmodium spp., Toxoplasma spp., and Trypanosoma spp. IVM has led to crucial findings on these parasite species, which are discussed in detail in this review.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasglow, UK
| | - Adéla Nacer
- Division of Bacteriology, National Institute for Biological Standards and Control, Medicines and Healthcare products Regulatory Agency, EN63QG, Potters Bar, UK
| | - Friedrich Frischknecht
- Parasitology-Centre for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| |
Collapse
|
45
|
Yap XZ, Lundie RJ, Beeson JG, O'Keeffe M. Dendritic Cell Responses and Function in Malaria. Front Immunol 2019; 10:357. [PMID: 30886619 PMCID: PMC6409297 DOI: 10.3389/fimmu.2019.00357] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/12/2019] [Indexed: 12/24/2022] Open
Abstract
Malaria remains a serious threat to global health. Sustained malaria control and, eventually, eradication will only be achieved with a broadly effective malaria vaccine. Yet a fundamental lack of knowledge about how antimalarial immunity is acquired has hindered vaccine development efforts to date. Understanding how malaria-causing parasites modulate the host immune system, specifically dendritic cells (DCs), key initiators of adaptive and vaccine antigen-based immune responses, is vital for effective vaccine design. This review comprehensively summarizes how exposure to Plasmodium spp. impacts human DC function in vivo and in vitro. We have highlighted the heterogeneity of the data observed in these studies, compared and critiqued the models used to generate our current understanding of DC function in malaria, and examined the mechanisms by which Plasmodium spp. mediate these effects. This review highlights potential research directions which could lead to improved efficacy of existing vaccines, and outlines novel targets for next-generation vaccine strategies to target malaria.
Collapse
Affiliation(s)
- Xi Zen Yap
- Burnet Institute, Melbourne, VIC, Australia.,Department of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Rachel J Lundie
- Burnet Institute, Melbourne, VIC, Australia.,Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia.,Department of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Parkville, VIC, Australia.,Department of Microbiology and Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Meredith O'Keeffe
- Burnet Institute, Melbourne, VIC, Australia.,Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
46
|
Walk J, Stok JE, Sauerwein RW. Can Patrolling Liver-Resident T Cells Control Human Malaria Parasite Development? Trends Immunol 2019; 40:186-196. [PMID: 30713008 DOI: 10.1016/j.it.2019.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/08/2019] [Accepted: 01/13/2019] [Indexed: 12/23/2022]
Abstract
Recently, a population of non-recirculating, tissue-resident memory CD8+ T cells has been identified; cells that seems to act as key sentinels for invading microorganisms with enhanced effector functions. In malaria, the liver represents the first site for parasite development before a definite infection is established in circulating red blood cells. Here, we discuss the evidence obtained from animal models on several diseases and hypothesize that liver-resident memory CD8+ T cells (hepatic TRM) play a critical role in providing protective liver-stage immunity against Plasmodium malaria parasites. Although observations in human malaria trials are limited to peripheral blood, we propose recommendations for the translation of some of these findings to human malaria research.
Collapse
Affiliation(s)
- Jona Walk
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Jorn E Stok
- University Medical Center Utrecht, PO Box 85500, Utrecht, The Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
47
|
Tyagi RK, Tandel N, Deshpande R, Engelman RW, Patel SD, Tyagi P. Humanized Mice Are Instrumental to the Study of Plasmodium falciparum Infection. Front Immunol 2018; 9:2550. [PMID: 30631319 PMCID: PMC6315153 DOI: 10.3389/fimmu.2018.02550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/17/2018] [Indexed: 02/05/2023] Open
Abstract
Research using humanized mice has advanced our knowledge and understanding of human haematopoiesis, non-adaptive and adaptive immunity, autoimmunity, infectious disease, cancer biology, and regenerative medicine. Challenges posed by the human-malaria parasite Plasmodium falciparum include its complex life cycle, the evolution of drug resistance against anti-malarials, poor diagnosis, and a lack of effective vaccines. Advancements in genetically engineered and immunodeficient mouse strains, have allowed for studies of the asexual blood stage, exoerythrocytic stage and the transition from liver-to-blood stage infection, in a single vertebrate host. This review discusses the process of "humanization" of various immunodeficient/transgenic strains and their contribution to translational biomedical research. Our work reviews the strategies employed to overcome the remaining-limitations of the developed human-mouse chimera(s).
Collapse
Affiliation(s)
- Rajeev K. Tyagi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Biomedical parasitology Unit, Institute Pasteur, Paris, France
- Department of Global Health, College of Public Health, University of South Florida, Tampa, FL, United States
| | - Nikunj Tandel
- Institute of Science, Nirma University, Ahmedabad, India
| | | | - Robert W. Engelman
- Department of Pediatrics, Pathology and Cell Biology, University of South Florida, Tampa, FL, United States
| | | | - Priyanka Tyagi
- Department of Basic and Applied Sciences, School of Engineering, GD Goenka University, Gurgaon, India
| |
Collapse
|
48
|
Glennon EKK, Dankwa S, Smith JD, Kaushansky A. Opportunities for Host-targeted Therapies for Malaria. Trends Parasitol 2018; 34:843-860. [PMID: 30122551 PMCID: PMC6168423 DOI: 10.1016/j.pt.2018.07.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 12/19/2022]
Abstract
Despite the recent successes of artemisinin-based antimalarial drugs, many still die from severe malaria, and eradication efforts are hindered by the limited drugs currently available to target transmissible gametocyte parasites and liver-resident dormant Plasmodium vivax hypnozoites. Host-targeted therapy is a new direction for infectious disease drug development and aims to interfere with host molecules, pathways, or networks that are required for infection or that contribute to disease. Recent advances in our understanding of host pathways involved in parasite development and pathogenic mechanisms in severe malaria could facilitate the development of host-targeted interventions against Plasmodium infection and malaria disease. This review discusses new opportunities for host-targeted therapeutics for malaria and the potential to harness drug polypharmacology to simultaneously target multiple host pathways using a single drug intervention.
Collapse
Affiliation(s)
- Elizabeth K K Glennon
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory, Box 357965, Seattle, WA 98195, USA; These authors made an equal contribution
| | - Selasi Dankwa
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; These authors made an equal contribution
| | - Joseph D Smith
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory, Box 357965, Seattle, WA 98195, USA
| | - Alexis Kaushansky
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory, Box 357965, Seattle, WA 98195, USA.
| |
Collapse
|