1
|
Goswami D, Patel H, Betz W, Armstrong J, Camargo N, Patil A, Chakravarty S, Murphy SC, Sim BKL, Vaughan AM, Hoffman SL, Kappe SH. A replication competent Plasmodium falciparum parasite completely attenuated by dual gene deletion. EMBO Mol Med 2024; 16:723-754. [PMID: 38514791 PMCID: PMC11018819 DOI: 10.1038/s44321-024-00057-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Vaccination with infectious Plasmodium falciparum (Pf) sporozoites (SPZ) administered with antimalarial drugs (PfSPZ-CVac), confers superior sterilizing protection against infection when compared to vaccination with replication-deficient, radiation-attenuated PfSPZ. However, the requirement for drug administration constitutes a major limitation for PfSPZ-CVac. To obviate this limitation, we generated late liver stage-arresting replication competent (LARC) parasites by deletion of the Mei2 and LINUP genes (mei2-/linup- or LARC2). We show that Plasmodium yoelii (Py) LARC2 sporozoites did not cause breakthrough blood stage infections and engendered durable sterilizing immunity against various infectious sporozoite challenges in diverse strains of mice. We next genetically engineered a PfLARC2 parasite strain that was devoid of extraneous DNA and produced cryopreserved PfSPZ-LARC2. PfSPZ-LARC2 liver stages replicated robustly in liver-humanized mice but displayed severe defects in late liver stage differentiation and did not form liver stage merozoites. This resulted in complete abrogation of parasite transition to viable blood stage infection. Therefore, PfSPZ-LARC2 is the next-generation vaccine strain expected to unite the safety profile of radiation-attenuated PfSPZ with the superior protective efficacy of PfSPZ-CVac.
Collapse
Affiliation(s)
- Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Hardik Patel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - William Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Janna Armstrong
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Asha Patil
- Sanaria Inc., 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | | | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - B Kim Lee Sim
- Sanaria Inc., 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Stefan Hi Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
2
|
Patel H, Minkah NK, Kumar S, Zanghi G, Schepis A, Goswami D, Armstrong J, Abatiyow BA, Betz W, Reynolds L, Camargo N, Sheikh AA, Kappe SHI. Malaria blood stage infection suppresses liver stage infection via host-induced interferons but not hepcidin. Nat Commun 2024; 15:2104. [PMID: 38453916 PMCID: PMC10920859 DOI: 10.1038/s41467-024-46270-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Abstract
Malaria-causing Plasmodium parasites first replicate as liver stages (LS), which then seed symptomatic blood stage (BS) infection. Emerging evidence suggests that these stages impact each other via perturbation of host responses, and this influences the outcome of natural infection. We sought to understand whether the parasite stage interplay would affect live-attenuated whole parasite vaccination, since the efficacy of whole parasite vaccines strongly correlates with their extend of development in the liver. We thus investigated the impact of BS infection on LS development of genetically attenuated and wildtype parasites in female rodent malaria models and observed that for both, LS infection suffered severe suppression during concurrent BS infection. Strikingly and in contrast to previously published studies, we find that the BS-induced iron-regulating hormone hepcidin is not mediating suppression of LS development. Instead, we demonstrate that BS-induced host interferons are the main mediators of LS developmental suppression. The type of interferon involved depended on the BS-causing parasite species. Our study provides important mechanistic insights into the BS-mediated suppression of LS development. This has direct implications for understanding the outcomes of live-attenuated Plasmodium parasite vaccination in malaria-endemic areas and might impact the epidemiology of natural malaria infection.
Collapse
Affiliation(s)
- Hardik Patel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nana K Minkah
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Gigliola Zanghi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Antonino Schepis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Janna Armstrong
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Biley A Abatiyow
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Will Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Laura Reynolds
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Amina A Sheikh
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
3
|
Cryopreservation of Plasmodium Sporozoites. Pathogens 2022; 11:pathogens11121487. [PMID: 36558821 PMCID: PMC9784981 DOI: 10.3390/pathogens11121487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Malaria is a deadly disease caused by the parasite, Plasmodium, and impacts the lives of millions of people around the world. Following inoculation into mammalian hosts by infected mosquitoes, the sporozoite stage of Plasmodium undergoes obligate development in the liver before infecting erythrocytes and causing clinical malaria. The most promising vaccine candidates for malaria rely on the use of attenuated live sporozoites to induce protective immune responses. The scope of widespread testing or clinical use of such vaccines is limited by the absence of efficient, reliable, or transparent strategies for the long-term preservation of live sporozoites. Here we outline a method to cryopreserve the sporozoites of various human and murine Plasmodium species. We found that the structural integrity, viability, and in vivo or in vitro infectiousness were conserved in the recovered cryopreserved sporozoites. Cryopreservation using our approach also retained the transgenic properties of sporozoites and immunization with cryopreserved radiation attenuated sporozoites (RAS) elicited strong immune responses. Our work offers a reliable protocol for the long-term storage and recovery of human and murine Plasmodium sporozoites and lays the groundwork for the widespread use of live sporozoites for research and clinical applications.
Collapse
|
4
|
Wilder BK, Vigdorovich V, Carbonetti S, Minkah N, Hertoghs N, Raappana A, Cardamone H, Oliver BG, Trakhimets O, Kumar S, Dambrauskas N, Arredondo SA, Camargo N, Seilie AM, Murphy SC, Kappe SHI, Sather DN. Anti-TRAP/SSP2 monoclonal antibodies can inhibit sporozoite infection and may enhance protection of anti-CSP monoclonal antibodies. NPJ Vaccines 2022; 7:58. [PMID: 35618791 PMCID: PMC9135708 DOI: 10.1038/s41541-022-00480-2] [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: 10/19/2021] [Accepted: 04/22/2022] [Indexed: 11/10/2022] Open
Abstract
Vaccine-induced sterilizing protection from infection by Plasmodium parasites, the pathogens that cause malaria, will be essential in the fight against malaria as it would prevent both malaria-related disease and transmission. Stopping the relatively small number of parasites injected by the mosquito before they can migrate from the skin to the liver is an attractive means to this goal. Antibody-eliciting vaccines have been used to pursue this objective by targeting the major parasite surface protein present during this stage, the circumsporozoite protein (CSP). While CSP-based vaccines have recently had encouraging success in disease reduction, this was only achieved with extremely high antibody titers and appeared less effective for a complete block of infection (i.e., sterile protection). While such disease reduction is important, these and other results indicate that strategies focusing on CSP alone may not achieve the high levels of sterile protection needed for malaria eradication. Here, we show that monoclonal antibodies (mAbs) recognizing another sporozoite protein, TRAP/SSP2, exhibit a range of inhibitory activity and that these mAbs may augment CSP-based protection despite conferring no sterile protection on their own. Therefore, pursuing a multivalent subunit vaccine immunization is a promising strategy for improving infection-blocking malaria vaccines.
Collapse
Affiliation(s)
- Brandon K Wilder
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Vladimir Vigdorovich
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sara Carbonetti
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nana Minkah
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nina Hertoghs
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Andrew Raappana
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Hayley Cardamone
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Brian G Oliver
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Olesya Trakhimets
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nicholas Dambrauskas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Silvia A Arredondo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Annette M Seilie
- Department of Laboratory Medicine and Pathology and Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology and Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
5
|
Steel RWJ, Vigdorovich V, Dambrauskas N, Wilder BK, Arredondo SA, Goswami D, Kumar S, Carbonetti S, Swearingen KE, Nguyen T, Betz W, Camargo N, Fisher BS, Soden J, Thomas H, Freeth J, Moritz RL, Noah Sather D, Kappe SHI. Platelet derived growth factor receptor β (PDGFRβ) is a host receptor for the human malaria parasite adhesin TRAP. Sci Rep 2021; 11:11328. [PMID: 34059712 PMCID: PMC8166973 DOI: 10.1038/s41598-021-90722-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/13/2021] [Indexed: 02/04/2023] Open
Abstract
Following their inoculation by the bite of an infected Anopheles mosquito, the malaria parasite sporozoite forms travel from the bite site in the skin into the bloodstream, which transports them to the liver. The thrombospondin-related anonymous protein (TRAP) is a type 1 transmembrane protein that is released from secretory organelles and relocalized on the sporozoite plasma membrane. TRAP is required for sporozoite motility and host infection, and its extracellular portion contains adhesive domains that are predicted to engage host receptors. Here, we identified the human platelet-derived growth factor receptor β (hPDGFRβ) as one such protein receptor. Deletion constructs showed that the von Willebrand factor type A and thrombospondin repeat domains of TRAP are both required for optimal binding to hPDGFRβ-expressing cells. We also demonstrate that this interaction is conserved in the human-infective parasite Plasmodium vivax, but not the rodent-infective parasite Plasmodium yoelii. We observed expression of hPDGFRβ mainly in cells associated with the vasculature suggesting that TRAP:hPDGFRβ interaction may play a role in the recognition of blood vessels by invading sporozoites.
Collapse
Affiliation(s)
- Ryan W J Steel
- Seattle Children's Research Institute, Seattle, WA, USA
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | | | | | - Brandon K Wilder
- Seattle Children's Research Institute, Seattle, WA, USA
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | | | | | - Sudhir Kumar
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | - Thao Nguyen
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Will Betz
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Jo Soden
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | - Helen Thomas
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | - Jim Freeth
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | | | - D Noah Sather
- Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Stefan H I Kappe
- Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
6
|
Ramaprasad A, Klaus S, Douvropoulou O, Culleton R, Pain A. Plasmodium vinckei genomes provide insights into the pan-genome and evolution of rodent malaria parasites. BMC Biol 2021; 19:69. [PMID: 33888092 PMCID: PMC8063448 DOI: 10.1186/s12915-021-00995-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/25/2021] [Indexed: 01/27/2023] Open
Abstract
Background Rodent malaria parasites (RMPs) serve as tractable tools to study malaria parasite biology and host-parasite-vector interactions. Among the four RMPs originally collected from wild thicket rats in sub-Saharan Central Africa and adapted to laboratory mice, Plasmodium vinckei is the most geographically widespread with isolates collected from five separate locations. However, there is a lack of extensive phenotype and genotype data associated with this species, thus hindering its use in experimental studies. Results We have generated a comprehensive genetic resource for P. vinckei comprising of five reference-quality genomes, one for each of its subspecies, blood-stage RNA sequencing data for five P. vinckei isolates, and genotypes and growth phenotypes for ten isolates. Additionally, we sequenced seven isolates of the RMP species Plasmodium chabaudi and Plasmodium yoelii, thus extending genotypic information for four additional subspecies enabling a re-evaluation of the genotypic diversity and evolutionary history of RMPs. The five subspecies of P. vinckei have diverged widely from their common ancestor and have undergone large-scale genome rearrangements. Comparing P. vinckei genotypes reveals region-specific selection pressures particularly on genes involved in mosquito transmission. Using phylogenetic analyses, we show that RMP multigene families have evolved differently across the vinckei and berghei groups of RMPs and that family-specific expansions in P. chabaudi and P. vinckei occurred in the common vinckei group ancestor prior to speciation. The erythrocyte membrane antigen 1 and fam-c families in particular show considerable expansions among the lowland forest-dwelling P. vinckei parasites. The subspecies from the highland forests of Katanga, P. v. vinckei, has a uniquely smaller genome, a reduced multigene family repertoire and is also amenable to transfection making it an ideal parasite for reverse genetics. We also show that P. vinckei parasites are amenable to genetic crosses. Conclusions Plasmodium vinckei isolates display a large degree of phenotypic and genotypic diversity and could serve as a resource to study parasite virulence and immunogenicity. Inclusion of P. vinckei genomes provide new insights into the evolution of RMPs and their multigene families. Amenability to genetic crossing and transfection make them also suitable for classical and functional genetics to study Plasmodium biology. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-00995-5.
Collapse
Affiliation(s)
- Abhinay Ramaprasad
- Pathogen Genomics Group, BESE Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia.,Malaria Unit, Department of Pathology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Present address: Malaria Biochemistry Laboratory, Francis Crick Institute, London, NW1 1AT, UK
| | - Severina Klaus
- Malaria Unit, Department of Pathology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Biomedical Sciences, University of Heidelberg, Heidelberg, Germany
| | - Olga Douvropoulou
- Pathogen Genomics Group, BESE Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Richard Culleton
- Malaria Unit, Department of Pathology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan. .,Division of Molecular Parasitology, Proteo-Science Center, Ehime University, 454 Shitsukawa, Toon, Ehime, 791-0295, Japan. .,Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| | - Arnab Pain
- Pathogen Genomics Group, BESE Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia. .,Center for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, N20 W10 Kita-ku, Sapporo, 001-0020, Japan.
| |
Collapse
|
7
|
Afifi MA. The Parasites Caught In-Action: Imaging at the Host-Parasite Interface. J Microsc Ultrastruct 2021; 9:1-6. [PMID: 33850705 PMCID: PMC8030542 DOI: 10.4103/jmau.jmau_1_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 01/02/2020] [Accepted: 01/21/2020] [Indexed: 11/24/2022] Open
Abstract
For many decades, scientists were unable to expose the invisible existence of the parasites in their living hosts, except by scarification and then dissection of the animal model. This process just demonstrates a dead parasite in a dead host. Using this approach, very limited information can be obtained concerning the dynamics of infection and the pathways utilized by the parasite to survive within a hostile host's environment. Introduction of ultra-high-speed imaging techniques, with a time domain of barely few microseconds or even less, has revolutionized the "in vivo dissection" of the parasites. Such methods provide platforms for imaging host-parasite interactions at diverse scales, down to the molecular level. These have complementary advantages and relative assets in investigating host-parasite interactions. Therefore, better elucidation of such interaction may require the usage of more than one approach. Precise in vivo quantification, of the parasite load within the host, and better insight into the kinetics of infection are the two main advantages of the novel imaging procedures. However, imaging parasite-host interplay is still a challenging approach due to many constraints related to the parasite biology, the tissue environment within which the parasites exist, and the logistic technical limitations. This review was planned to assist better understanding of how much the new imaging techniques impacted the recent advances in parasite biology, especially the immunobiology of protozoan parasites.
Collapse
Affiliation(s)
- Mohammed A. Afifi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Parasitology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
8
|
Vancomycin improves Plasmodium yoelii malaria parasite in vitro liver stage cultures by controlling Elizabethkingia anophelis, a bacterium in the microbiome of lab-reared Anopheles mosquitoes. Mol Biochem Parasitol 2020; 237:111279. [PMID: 32360511 DOI: 10.1016/j.molbiopara.2020.111279] [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: 02/23/2020] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 11/23/2022]
Abstract
Studies of Plasmodium sporozoites and liver stages require dissection of Anopheles mosquitoes to obtain sporozoites for experiments. Sporozoites from the rodent parasite P. yoelii are routinely used to infect hepatocytes for liver stage culture, but sometimes these cultures become contaminated. Using standard microbiological techniques, a single colony type of Gram-negative rod-shaped bacteria was isolated from contaminated cultures. Mass spectrometry and sequencing of the bacterial 16S ribosomal RNA gene identified the contaminant as Elizabethkingia spp. Based on sequence comparison and published studies of the Anopheles microbiome, the best match was E. anophelis. Culture contamination was not ameliorated by density gradient purification of sporozoites. However, the addition of vancomycin to the culture media consistently reduced contamination and improved culture outcomes as measured by liver stage parasite size. Thus, mosquito salivary gland-derived E. anophelis is identified a potential contaminant of Plasmodium liver stage cultures that can be mitigated by the addition of antibiotics.
Collapse
|
9
|
Glennon EKK, Austin LS, Arang N, Kain HS, Mast FD, Vijayan K, Aitchison JD, Kappe SHI, Kaushansky A. Alterations in Phosphorylation of Hepatocyte Ribosomal Protein S6 Control Plasmodium Liver Stage Infection. Cell Rep 2020; 26:3391-3399.e4. [PMID: 30893610 DOI: 10.1016/j.celrep.2019.02.085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 10/11/2018] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
Plasmodium parasites are highly selective when infecting hepatocytes and induce many changes within the host cell upon infection. While several host cell factors have been identified that are important for liver infection, our understanding of what facilitates the maintenance of infection remains incomplete. Here, we describe a role for phosphorylated ribosomal protein S6 (Ser235/236) (p-RPS6) in Plasmodium yoelii-infected hepatocytes. Blocking RPS6 phosphorylation prior to infection decreases the number of liver stage parasites within 24 h. Infected hepatocytes exhibit elevated levels of p-RPS6 while simultaneously abrogating the induction of phosphorylation of RPS6 in response to insulin stimulation. This is in contrast with the regulation of p-RPS6 by Toxoplasma gondii, which elevates levels of p-RPS6 after infection but does not alter the response to insulin. Our data support a model in which RPS6 phosphorylation is uncoupled from canonical regulators in Plasmodium-infected hepatocytes and is relied on by the parasite to maintain infection.
Collapse
Affiliation(s)
- Elizabeth K K Glennon
- Center for Infectious Disease Research, Seattle, WA 98109, USA; Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Seattle, WA 98109, USA
| | - Laura S Austin
- Center for Infectious Disease Research, Seattle, WA 98109, USA
| | - Nadia Arang
- Center for Infectious Disease Research, Seattle, WA 98109, USA
| | - Heather S Kain
- Center for Infectious Disease Research, Seattle, WA 98109, USA
| | - Fred D Mast
- Center for Infectious Disease Research, Seattle, WA 98109, USA; Seattle Children's Research Institute, Seattle, WA 98109, USA; Institute for Systems Biology, Seattle, WA 98109, USA
| | - Kamalakannan Vijayan
- Center for Infectious Disease Research, Seattle, WA 98109, USA; Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - John D Aitchison
- Center for Infectious Disease Research, Seattle, WA 98109, USA; Seattle Children's Research Institute, Seattle, WA 98109, USA; Institute for Systems Biology, Seattle, WA 98109, USA
| | - Stefan H I Kappe
- Center for Infectious Disease Research, Seattle, WA 98109, USA; Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Seattle, WA 98109, USA
| | - Alexis Kaushansky
- Center for Infectious Disease Research, Seattle, WA 98109, USA; Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
10
|
Coombs RS, Blank ML, English ED, Adomako-Ankomah Y, Urama ICS, Martin AT, Yarovinsky F, Boyle JP. Immediate Interferon Gamma Induction Determines Murine Host Compatibility Differences between Toxoplasma gondii and Neospora caninum. Infect Immun 2020; 88:e00027-20. [PMID: 32014892 PMCID: PMC7093116 DOI: 10.1128/iai.00027-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/24/2022] Open
Abstract
Rodents are critical for the transmission of Toxoplasma gondii to the definitive feline host via predation, and this relationship has been extensively studied as a model for immune responses to parasites. Neospora caninum is a closely related coccidian parasite of ruminants and canines but is not naturally transmitted by rodents. We compared mouse innate immune responses to N. caninum and T. gondii and found marked differences in cytokine levels and parasite growth kinetics during the first 24 h postinfection (hpi). N. caninum-infected mice produced significantly higher levels of interleukin-12 (IL-12) and interferon gamma (IFN-γ) by as early as 4 hpi, but the level of IFN-γ was significantly lower or undetectable in T. gondii-infected mice during the first 24 hpi. "Immediate" IFN-γ and IL-12p40 production was not detected in MyD88-/- mice. However, unlike IL-12p40-/- and IFN-γ-/- mice, MyD88-/- mice survived N. caninum infections at the dose used in this study. Serial measures of parasite burden showed that MyD88-/- mice were more susceptible to N. caninum infections than wild-type (WT) mice, and control of parasite burdens correlated with a pulse of serum IFN-γ at 3 to 4 days postinfection in the absence of detectable IL-12. Immediate IFN-γ was partially dependent on the T. gondii mouse profilin receptor Toll-like receptor 11 (TLR11), but the ectopic expression of N. caninum profilin in T. gondii had no impact on early IFN-γ production or parasite proliferation. Our data indicate that T. gondii is capable of evading host detection during the first hours after infection, while N. caninum is not, and this is likely due to the early MyD88-dependent recognition of ligands other than profilin.
Collapse
Affiliation(s)
- Rachel S Coombs
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew L Blank
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Elizabeth D English
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yaw Adomako-Ankomah
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Andrew T Martin
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Felix Yarovinsky
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Jon P Boyle
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
11
|
Innate immunity limits protective adaptive immune responses against pre-erythrocytic malaria parasites. Nat Commun 2019; 10:3950. [PMID: 31477704 PMCID: PMC6718385 DOI: 10.1038/s41467-019-11819-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
Immunization with attenuated whole Plasmodium sporozoites constitutes a promising vaccination strategy. Compared to replication-deficient parasites, immunization with replication-competent parasites confers better protection and also induces a type I IFN (IFN-1) response, but whether this IFN-1 response has beneficial or adverse effects on vaccine-induced adaptive immunity is not known. Here, we show that IFN-1 signaling-deficient mice immunized with replication-competent sporozoites exhibit superior protection against infection. This correlates with superior CD8 T cell memory including reduced expression of the exhaustion markers PD-1 and LAG-3 on these cells and increased numbers of memory CD8 T cells in the liver. Moreover, the adoptive transfer of memory CD8 T cells from the livers of previously immunized IFN-1 signaling-deficient mice confers greater protection against liver stage parasites. However, the detrimental role of IFN-1 signaling is not CD8 T cell intrinsic. Together, our data demonstrate that liver stage-engendered IFN-1 signaling impairs hepatic CD8 T cell memory via a CD8 T cell-extrinsic mechanism. Here, Minkah et al. show that, while immunization with replication-competent Plasmodium parasites can confer sterile protection against infection, it also induces a type I interferon response that adversely affects anti-malaria immunity by affecting numbers of protective hepatic CD8 T cells and CD8 T cell function.
Collapse
|
12
|
Smilkstein MJ, Pou S, Krollenbrock A, Bleyle LA, Dodean RA, Frueh L, Hinrichs DJ, Li Y, Martinson T, Munar MY, Winter RW, Bruzual I, Whiteside S, Nilsen A, Koop DR, Kelly JX, Kappe SHI, Wilder BK, Riscoe MK. ELQ-331 as a prototype for extremely durable chemoprotection against malaria. Malar J 2019; 18:291. [PMID: 31455339 PMCID: PMC6712883 DOI: 10.1186/s12936-019-2921-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/17/2019] [Indexed: 12/02/2022] Open
Abstract
Background The potential benefits of long-acting injectable chemoprotection (LAI-C) against malaria have been recently recognized, prompting a call for suitable candidate drugs to help meet this need. On the basis of its known pharmacodynamic and pharmacokinetic profiles after oral dosing, ELQ-331, a prodrug of the parasite mitochondrial electron transport inhibitor ELQ-300, was selected for study of pharmacokinetics and efficacy as LAI-C in mice. Methods Four trials were conducted in which mice were injected with a single intramuscular dose of ELQ-331 or other ELQ-300 prodrugs in sesame oil with 1.2% benzyl alcohol; the ELQ-300 content of the doses ranged from 2.5 to 30 mg/kg. Initial blood stage challenges with Plasmodium yoelii were used to establish the model, but the definitive study measure of efficacy was outcome after sporozoite challenge with a luciferase-expressing P. yoelii, assessed by whole-body live animal imaging. Snapshot determinations of plasma ELQ-300 concentration ([ELQ-300]) were made after all prodrug injections; after the highest dose of ELQ-331 (equivalent to 30 mg/kg ELQ-300), both [ELQ-331] and [ELQ-300] were measured at a series of timepoints from 6 h to 5½ months after injection. Results A single intramuscular injection of ELQ-331 outperformed four other ELQ-300 prodrugs and, at a dose equivalent to 30 mg/kg ELQ-300, protected mice against challenge with P. yoelii sporozoites for at least 4½ months. Pharmacokinetic evaluation revealed rapid and essentially complete conversion of ELQ-331 to ELQ-300, a rapidly achieved (< 6 h) and sustained (4–5 months) effective plasma ELQ-300 concentration, maximum ELQ-300 concentrations far below the estimated threshold for toxicity, and a distinctive ELQ-300 concentration versus time profile. Pharmacokinetic modeling indicates a high-capacity, slow-exchange tissue compartment which serves to accumulate and then slowly redistribute ELQ-300 into blood, and this property facilitates an extremely long period during which ELQ-300 concentration is sustained above a minimum fully-protective threshold (60–80 nM). Conclusions Extrapolation of these results to humans predicts that ELQ-331 should be capable of meeting and far-exceeding currently published duration-of-effect goals for anti-malarial LAI-C. Furthermore, the distinctive pharmacokinetic profile of ELQ-300 after treatment with ELQ-331 may facilitate durable protection and enable protection for far longer than 3 months. These findings suggest that ELQ-331 warrants consideration as a leading prototype for LAI-C.
Collapse
Affiliation(s)
- Martin J Smilkstein
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA.
| | - Sovitj Pou
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - Alina Krollenbrock
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Lisa A Bleyle
- Bioanalytical Shared Resource Core Pharmacokinetics, Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, L334, Portland, OR, 97239, USA
| | - Rozalia A Dodean
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA.,Department of Chemistry, Portland State University, PO Box 751, Portland, OR, 97207, USA
| | - Lisa Frueh
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - David J Hinrichs
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - Yuexin Li
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - Thomas Martinson
- Vaccine & Gene Therapy Institute (VGTI), Oregon Health and Science University (West Campus), 505 NW 185th Avenue, #1, Beaverton, OR, 97006, USA
| | - Myrna Y Munar
- Oregon State University/Oregon Health and Science University College of Pharmacy, 2730 SW Moody Avenue, CL5CP, Portland, OR, 97201, USA
| | - Rolf W Winter
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA.,Department of Chemistry, Portland State University, PO Box 751, Portland, OR, 97207, USA
| | - Igor Bruzual
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - Samantha Whiteside
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Ave N., Suite 500, Seattle, WA, USA
| | - Aaron Nilsen
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - Dennis R Koop
- Bioanalytical Shared Resource Core Pharmacokinetics, Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, L334, Portland, OR, 97239, USA
| | - Jane X Kelly
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA.,Department of Chemistry, Portland State University, PO Box 751, Portland, OR, 97207, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Ave N., Suite 500, Seattle, WA, USA
| | - Brandon K Wilder
- Vaccine & Gene Therapy Institute (VGTI), Oregon Health and Science University (West Campus), 505 NW 185th Avenue, #1, Beaverton, OR, 97006, USA
| | - Michael K Riscoe
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| |
Collapse
|
13
|
Sun Y, Wang S, Yang H, Wu J, Li S, Qiao G, Wang S, Wang X, Zhou X, Osada T, Hobeika A, Morse MA, Ren J, Lyerly HK. Impact of synchronized anti-PD-1 with Ad-CEA vaccination on inhibition of colon cancer growth. Immunotherapy 2019; 11:953-966. [PMID: 31192764 DOI: 10.2217/imt-2019-0055] [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] [Indexed: 12/12/2022] Open
Abstract
Aim: The purpose of this study was to determine whether addition of anti-PD-1 antibody increased the immunogenicity and anti-tumor activity of Ad-CEA vaccination in a murine model of colon cancer. Methods: Ad-CEA was administered prior to implantation of MC-38-CEA cells followed by administration of anti-PD-1 antibody. CEA-specific T-cell responses were measured by flow cytometry and ELISPOT. Dynamic co-culture of splenocytes with tumor cells was conducted to analyze anti-tumor activities. Tumor infiltration by lymphocytes was measured by IHC. Tumor volume and overall survival were also recorded. Results: Ad-CEA combined with anti-PD-1 antibody showed greater anti-tumor activity compared with either alone. The combination also increased T-cell infiltration but decreased Tregs. Conclusion: Combining Ad-CEA vaccination with anti-PD-1 antibody enhanced anti-tumor activity and immune responses.
Collapse
Affiliation(s)
- Yuanyuan Sun
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Suya Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Hainan Yang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Jiangping Wu
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Sha Li
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Guoliang Qiao
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Shuo Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xinna Zhou
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Takuya Osada
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Michael A Morse
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Jun Ren
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.,Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
14
|
Updating the modified Thompson test by using whole-body bioluminescence imaging to replace traditional efficacy testing in experimental models of murine malaria. Malar J 2019; 18:38. [PMID: 30767768 PMCID: PMC6376706 DOI: 10.1186/s12936-019-2661-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 01/19/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rodent malaria models are extensively used to predict treatment outcomes in human infections. There is a constant need to improve and refine these models by innovating ways to apply new scientific findings and cutting edge technologies. In addition, and in accordance with the three R's of animal use in research, in vivo studies should be constantly refined to avoid unnecessary pain and distress to the experimental animals by using preemptive euthanasia as soon as the main scientific study objective has been accomplished. METHODS The new methodology described in this manuscript uses the whole-body bioluminescence signal emitted by transgenic, luciferase-expressing Plasmodium berghei parasites to assess the parasite load predicted parasitaemia (PLPP) in drug and control treated female ICR-CD1 mice infected with 1 × 105 luciferase-expressing P. berghei (ANKA strain) infected erythrocytes. This methodology can replace other time-consuming and expensive methods that are routinely used to measure parasitaemia in infected animals, such as Giemsa-stained thin blood smears and flow cytometry. RESULTS There is a good correlation between whole-body bioluminescence signal and parasitaemia measured using Giemsa-stained thin blood smears and flow cytometry respectively in donor and study mice in the modified Thompson test. The algebraic formulas which represent these correlations can be successfully used to assess PLPP in donor and study mice. In addition, the new methodology can pinpoint sick animals 2-8 days before they would have been otherwise diagnosed based on behavioural or any other signs of malaria disease. CONCLUSIONS The new method for predicting parasitaemia in the modified Thompson test is simple, precise, objective, and minimizes false positive results that can lead to the premature removal of animals from study. Furthermore, from the animal welfare perspective of replace, reduce, and refine, this new method facilitates early removal of sick animals from study as soon as the study objective has been achieved, in many cases well before the clinical signs of disease are present.
Collapse
|
15
|
A human monoclonal antibody prevents malaria infection by targeting a new site of vulnerability on the parasite. Nat Med 2018; 24:408-416. [PMID: 29554083 PMCID: PMC5893371 DOI: 10.1038/nm.4512] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 01/07/2023]
Abstract
Development of a highly effective vaccine or antibodies for the prevention and ultimately elimination of malaria is urgently needed. Here we report the isolation of a number of human monoclonal antibodies directed against the Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP) from several subjects immunized with an attenuated Pf whole-sporozoite (SPZ) vaccine (Sanaria PfSPZ Vaccine). Passive transfer of one of these antibodies, monoclonal antibody CIS43, conferred high-level, sterile protection in two different mouse models of malaria infection. The affinity and stoichiometry of CIS43 binding to PfCSP indicate that there are two sequential multivalent binding events encompassing the repeat domain. The first binding event is to a unique 'junctional' epitope positioned between the N terminus and the central repeat domain of PfCSP. Moreover, CIS43 prevented proteolytic cleavage of PfCSP on PfSPZ. Analysis of crystal structures of the CIS43 antigen-binding fragment in complex with the junctional epitope determined the molecular interactions of binding, revealed the epitope's conformational flexibility and defined Asn-Pro-Asn (NPN) as the structural repeat motif. The demonstration that CIS43 is highly effective for passive prevention of malaria has potential application for use in travelers, military personnel and elimination campaigns and identifies a new and conserved site of vulnerability on PfCSP for next-generation rational vaccine design.
Collapse
|
16
|
Bosson-Vanga H, Franetich JF, Soulard V, Sossau D, Tefit M, Kane B, Vaillant JC, Borrmann S, Müller O, Dereuddre-Bosquet N, Le Grand R, Silvie O, Mazier D. Differential activity of methylene blue against erythrocytic and hepatic stages of Plasmodium. Malar J 2018; 17:143. [PMID: 29615050 PMCID: PMC5883292 DOI: 10.1186/s12936-018-2300-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/26/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND In the context of malaria elimination/eradication, drugs that are effective against the different developmental stages of the parasite are highly desirable. The oldest synthetic anti-malarial drug, the thiazine dye methylene blue (MB), is known for its activity against Plasmodium blood stages, including gametocytes. The aim of the present study was to investigate a possible effect of MB against malaria parasite liver stages. METHODS MB activity was investigated using both in vitro and in vivo models. In vitro assays consisted of testing MB activity on Plasmodium falciparum, Plasmodium cynomolgi and Plasmodium yoelii parasites in human, simian or murine primary hepatocytes, respectively. MB in vivo activity was evaluated using intravital imaging in BALB/c mice infected with a transgenic bioluminescent P. yoelii parasite line. The transmission-blocking activity of MB was also addressed using mosquitoes fed on MB-treated mice. RESULTS MB shows no activity on Plasmodium liver stages, including hypnozoites, in vitro in primary hepatocytes. In BALB/c mice, MB has moderate effect on P. yoelii hepatic development but is highly effective against blood stage growth. MB is active against gametocytes and abrogates parasite transmission from mice to mosquitoes. CONCLUSION While confirming activity of MB against both sexual and asexual blood stages, the results indicate that MB has only little activity on the development of the hepatic stages of malaria parasites.
Collapse
Affiliation(s)
- Henriette Bosson-Vanga
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, U1135, ERL8255, CIMI-Paris, F-75013, PARIS, France. .,Département de Parasitologie-Mycologie, UFR des Sciences Pharmaceutiques et Biologiques, Université Félix Houphouët Boigny, Abidjan, Côte d'Ivoire.
| | - Jean-François Franetich
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, U1135, ERL8255, CIMI-Paris, F-75013, PARIS, France
| | - Valérie Soulard
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, U1135, ERL8255, CIMI-Paris, F-75013, PARIS, France
| | - Daniel Sossau
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, U1135, ERL8255, CIMI-Paris, F-75013, PARIS, France.,Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Maurel Tefit
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, U1135, ERL8255, CIMI-Paris, F-75013, PARIS, France
| | - Bocar Kane
- UPMC, UMS28, 105 Bd de l'hôpital, 75013, Paris, France
| | - Jean-Christophe Vaillant
- Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, 83 Bd de l'hôpital, 75013, Paris, France
| | - Steffen Borrmann
- German Center for Infection Research (DZIF), Tübingen, Germany.,Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Olaf Müller
- Institute of Public Health, Medical School, Ruprecht-Karls-University, Heidelberg, Germany
| | - Nathalie Dereuddre-Bosquet
- CEA, INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Université Paris Sud 11, Fontenay-aux-Roses, France
| | - Roger Le Grand
- CEA, INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Université Paris Sud 11, Fontenay-aux-Roses, France
| | - Olivier Silvie
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, U1135, ERL8255, CIMI-Paris, F-75013, PARIS, France
| | - Dominique Mazier
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, U1135, ERL8255, CIMI-Paris, F-75013, PARIS, France. .,Service de Parasitologie-Mycologie, Centre National de Référence du Paludisme, AP-HP, Groupe Hospitalier Pitié Salpêtrière, 83 Bd de l'hôpital, 75013, PARIS, France.
| |
Collapse
|
17
|
Avci P, Karimi M, Sadasivam M, Antunes-Melo WC, Carrasco E, Hamblin MR. In-vivo monitoring of infectious diseases in living animals using bioluminescence imaging. Virulence 2017; 9:28-63. [PMID: 28960132 PMCID: PMC6067836 DOI: 10.1080/21505594.2017.1371897] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Traditional methods of localizing and quantifying the presence of pathogenic microorganisms in living experimental animal models of infections have mostly relied on sacrificing the animals, dissociating the tissue and counting the number of colony forming units. However, the discovery of several varieties of the light producing enzyme, luciferase, and the genetic engineering of bacteria, fungi, parasites and mice to make them emit light, either after administration of the luciferase substrate, or in the case of the bacterial lux operon without any exogenous substrate, has provided a new alternative. Dedicated bioluminescence imaging (BLI) cameras can record the light emitted from living animals in real time allowing non-invasive, longitudinal monitoring of the anatomical location and growth of infectious microorganisms as measured by strength of the BLI signal. BLI technology has been used to follow bacterial infections in traumatic skin wounds and burns, osteomyelitis, infections in intestines, Mycobacterial infections, otitis media, lung infections, biofilm and endodontic infections and meningitis. Fungi that have been engineered to be bioluminescent have been used to study infections caused by yeasts (Candida) and by filamentous fungi. Parasitic infections caused by malaria, Leishmania, trypanosomes and toxoplasma have all been monitored by BLI. Viruses such as vaccinia, herpes simplex, hepatitis B and C and influenza, have been studied using BLI. This rapidly growing technology is expected to continue to provide much useful information, while drastically reducing the numbers of animals needed in experimental studies.
Collapse
Affiliation(s)
- Pinar Avci
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,b Department of Dermatology , Harvard Medical School , Boston , MA , USA
| | - Mahdi Karimi
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,c Department of Medical Nanotechnology , School of Advanced Technologies in Medicine, Iran University of Medical Sciences , Tehran , Iran.,d Cellular and Molecular Research Center, Iran University of Medical Sciences , Tehran , Iran
| | - Magesh Sadasivam
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,e Amity Institute of Nanotechnology, Amity University Uttar Pradesh , Noida , India
| | - Wanessa C Antunes-Melo
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,f University of Sao Paulo , Sao Carlos-SP , Brazil
| | - Elisa Carrasco
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,g Department of Biosciences , Durham University , Durham , United Kingdom
| | - Michael R Hamblin
- a Wellman Center for Photomedicine, Massachusetts General Hospital , Boston , MA , USA.,b Department of Dermatology , Harvard Medical School , Boston , MA , USA.,h Harvard-MIT Division of Health Sciences and Technology , Cambridge , MA , USA
| |
Collapse
|
18
|
Blood Stage Malaria Disrupts Humoral Immunity to the Pre-erythrocytic Stage Circumsporozoite Protein. Cell Rep 2017; 17:3193-3205. [PMID: 28009289 DOI: 10.1016/j.celrep.2016.11.060] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 09/18/2016] [Accepted: 11/18/2016] [Indexed: 11/21/2022] Open
Abstract
Many current malaria vaccines target the pre-erythrocytic stage of infection in the liver. However, in malaria-endemic regions, increased blood stage exposure is associated with decreased vaccine efficacy, thereby challenging current vaccine efforts. We hypothesized that pre-erythrocytic humoral immunity is directly disrupted by blood stage infection. To investigate this possibility, we used Plasmodium-antigen tetramers to analyze B cells after infection with either late liver stage arresting parasites or wild-type parasites that progress to the blood stage. Our data demonstrate that immunoglobulin G (IgG) antibodies against the pre-erythrocytic antigen, circumsporozoite protein (CSP), are generated only in response to the attenuated, but not the wild-type, infection. Further analyses revealed that blood stage malaria inhibits CSP-specific germinal center B cell differentiation and modulates chemokine expression. This results in aberrant memory formation and the loss of a rapid secondary B cell response. These data highlight how immunization with attenuated parasites may drive optimal immunity to malaria.
Collapse
|
19
|
Sack BK, Mikolajczak SA, Fishbaugher M, Vaughan AM, Flannery EL, Nguyen T, Betz W, Jane Navarro M, Foquet L, Steel RWJ, Billman ZP, Murphy SC, Hoffman SL, Chakravarty S, Sim BKL, Behet M, Reuling IJ, Walk J, Scholzen A, Sauerwein RW, Ishizuka AS, Flynn B, Seder RA, Kappe SHI. Humoral protection against mosquito bite-transmitted Plasmodium falciparum infection in humanized mice. NPJ Vaccines 2017; 2:27. [PMID: 29263882 PMCID: PMC5634440 DOI: 10.1038/s41541-017-0028-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 07/25/2017] [Accepted: 09/07/2017] [Indexed: 01/05/2023] Open
Abstract
A malaria vaccine that prevents infection will be an important new tool in continued efforts of malaria elimination, and such vaccines are under intense development for the major human malaria parasite Plasmodium falciparum (Pf). Antibodies elicited by vaccines can block the initial phases of parasite infection when sporozoites are deposited into the skin by mosquito bite and then target the liver for further development. However, there are currently no standardized in vivo preclinical models that can measure the inhibitory activity of antibody specificities against Pf sporozoite infection via mosquito bite. Here, we use human liver-chimeric mice as a challenge model to assess prevention of natural Pf sporozoite infection by antibodies. We demonstrate that these mice are consistently infected with Pf by mosquito bite and that this challenge can be combined with passive transfer of either monoclonal antibodies or polyclonal human IgG from immune serum to measure antibody-mediated blocking of parasite infection using bioluminescent imaging. This methodology is useful to down-select functional antibodies and to investigate mechanisms or immune correlates of protection in clinical trials, thereby informing rational vaccine optimization.
Collapse
Affiliation(s)
| | | | | | | | | | - Thao Nguyen
- Center for Infectious Disease Research, Seattle, WA USA
| | - Will Betz
- Center for Infectious Disease Research, Seattle, WA USA
| | | | - Lander Foquet
- Center for Infectious Disease Research, Seattle, WA USA
| | | | - Zachary P. Billman
- Departments of Laboratory Medicine and Microbiology and the Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA USA
| | - Sean C. Murphy
- Center for Infectious Disease Research, Seattle, WA USA
- Departments of Laboratory Medicine and Microbiology and the Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA USA
| | | | | | | | | | | | - Jona Walk
- Radboud University, Nijmegen, The Netherlands
| | | | | | | | | | | | - Stefan H. I. Kappe
- Center for Infectious Disease Research, Seattle, WA USA
- Department of Global Health, University of Washington, Seattle, WA USA
| |
Collapse
|
20
|
Othman AS, Marin-Mogollon C, Salman AM, Franke-Fayard BM, Janse CJ, Khan SM. The use of transgenic parasites in malaria vaccine research. Expert Rev Vaccines 2017; 16:1-13. [DOI: 10.1080/14760584.2017.1333426] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ahmad Syibli Othman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Catherin Marin-Mogollon
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | | | - Blandine M. Franke-Fayard
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Chris J. Janse
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Shahid M. Khan
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| |
Collapse
|
21
|
Burrows JN, Duparc S, Gutteridge WE, Hooft van Huijsduijnen R, Kaszubska W, Macintyre F, Mazzuri S, Möhrle JJ, Wells TNC. New developments in anti-malarial target candidate and product profiles. Malar J 2017; 16:26. [PMID: 28086874 PMCID: PMC5237200 DOI: 10.1186/s12936-016-1675-x] [Citation(s) in RCA: 320] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/30/2016] [Indexed: 11/10/2022] Open
Abstract
A decade of discovery and development of new anti-malarial medicines has led to a renewed focus on malaria elimination and eradication. Changes in the way new anti-malarial drugs are discovered and developed have led to a dramatic increase in the number and diversity of new molecules presently in pre-clinical and early clinical development. The twin challenges faced can be summarized by multi-drug resistant malaria from the Greater Mekong Sub-region, and the need to provide simplified medicines. This review lists changes in anti-malarial target candidate and target product profiles over the last 4 years. As well as new medicines to treat disease and prevent transmission, there has been increased focus on the longer term goal of finding new medicines for chemoprotection, potentially with long-acting molecules, or parenteral formulations. Other gaps in the malaria armamentarium, such as drugs to treat severe malaria and endectocides (that kill mosquitoes which feed on people who have taken the drug), are defined here. Ultimately the elimination of malaria requires medicines that are safe and well-tolerated to be used in vulnerable populations: in pregnancy, especially the first trimester, and in those suffering from malnutrition or co-infection with other pathogens. These updates reflect the maturing of an understanding of the key challenges in producing the next generation of medicines to control, eliminate and ultimately eradicate malaria.
Collapse
Affiliation(s)
- Jeremy N Burrows
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | - Stephan Duparc
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | | | | | - Wiweka Kaszubska
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | - Fiona Macintyre
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | | | - Jörg J Möhrle
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | - Timothy N C Wells
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland.
| |
Collapse
|
22
|
Steel RW, Kappe SH, Sack BK. An expanding toolkit for preclinical pre-erythrocytic malaria vaccine development: bridging traditional mouse malaria models and human trials. Future Microbiol 2016; 11:1563-1579. [PMID: 27855488 DOI: 10.2217/fmb-2016-0077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Malaria remains a significant public health burden with 214 million new infections and over 400,000 deaths in 2015. Elucidating relevant Plasmodium parasite biology can lead to the identification of novel ways to control and ultimately eliminate the parasite within geographic areas. Particularly, the development of an effective vaccine that targets the clinically silent pre-erythrocytic stages of infection would significantly augment existing malaria elimination tools by preventing both the onset of blood-stage infection/disease as well as spread of the parasite through mosquito transmission. In this Perspective, we discuss the role of small animal models in pre-erythrocytic stage vaccine development, highlighting how human liver-chimeric and human immune system mice are emerging as valuable components of these efforts.
Collapse
Affiliation(s)
- Ryan Wj Steel
- Center for Infectious Disease Research, Formerly Seattle Biomedical Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA 98109, USA
| | - Stefan Hi Kappe
- Center for Infectious Disease Research, Formerly Seattle Biomedical Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA 98109, USA
| | - Brandon K Sack
- Center for Infectious Disease Research, Formerly Seattle Biomedical Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA 98109, USA
| |
Collapse
|
23
|
SPECT/CT analysis of splenic function in genistein-treated malaria-infected mice. Exp Parasitol 2016; 170:10-15. [PMID: 27585499 DOI: 10.1016/j.exppara.2016.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 08/25/2016] [Accepted: 08/27/2016] [Indexed: 01/24/2023]
Abstract
Spleen traps malaria-infected red blood cells, thereby leading to splenomegaly. Splenomegaly induces impairment in splenic function, i.e., rupture. Therefore, splenomegaly inhibition is required to protect the spleen. In our previous study, genistein was found to have an influence on malaria-induced splenomegaly. However, the effect of genistein in malaria-induced splenomegaly, especially on the function of spleen, has not been fully investigated. In this study, hematoxylin and eosin (H&E) staining images show that genistein partially prevents malaria-induced architectural disruption of spleen. In addition, genistein decreases transgenic Plasmodium parasites accumulation in the spleen. Genistein treatment can protect splenic function from impairment caused by malaria infection. To examine the functions of malaria-infected spleen, we employed single-photon emission computed tomography/computed tomography (SPECT/CT) technology. Red blood cells are specifically radiolabeled with Technetium-99m pertechnetate (99mTcO4-) and trapped inside the spleen. The standardized uptake values (SUVs) in the spleen of infected mice are higher than those of naive and genistein-treated mice. However, genistein reduces the malaria-induced trapping capacity of spleen for heat-damaged radiolabeled RBCs, while exhibiting a protective effect against malaria. Considering these results, we suggested that genistein could be effectively used in combination therapy for malaria-induced splenic impairment.
Collapse
|
24
|
Billman ZP, Seilie AM, Murphy SC. Purification of Plasmodium Sporozoites Enhances Parasite-Specific CD8+ T Cell Responses. Infect Immun 2016; 84:2233-2242. [PMID: 27217420 PMCID: PMC4962643 DOI: 10.1128/iai.01439-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 05/14/2016] [Indexed: 01/26/2023] Open
Abstract
Malaria infection caused by Plasmodium parasites continues to cause enormous morbidity and mortality in areas where it is endemic, and there is no licensed vaccine capable of inducing sterile protection. Hyperimmunization with attenuated whole sporozoites can induce sterile protective immune responses targeting preerythrocytic antigens. Most animal models of hyperimmunization rely on sporozoites dissected from mosquito salivary glands and injected without further purification. In BALB/c mice, repeated small doses of P. yoelii sporozoites progressively expand the population of sporozoite-specific CD8(+) T cells. In this study, large secondary doses of unpurified sporozoites unexpectedly led to contraction of sporozoite-specific CD8(+) T cell responses in sporozoite-primed mice. While sporozoite-primed CD8(+) T cells alternatively can be expanded by secondary exposure to Listeria monocytogenes expressing recombinant Plasmodium antigens, such expansion was potently inhibited by coinjection of large doses of unpurified sporozoites and by uninfected salivary glands alone. Purification of sporozoites away from mosquito salivary gland debris by density gradient centrifugation eliminated salivary gland-associated inhibition. Thus, the inhibitory effect appears to be due to exposure to uninfected mosquito salivary glands rather than sporozoites. To further assess the effect of salivary gland exposure on later sporozoite vaccinations, mice were immunized with uninfected salivary glands from a single mosquito. Compared to naive mice, salivary gland presensitization reduced subsequent liver burdens by 71%. These data show that a component(s) in mosquito salivary glands reduces liver infection, thereby limiting antigen dose and contributing to lower-magnitude T cell responses. These findings suggest that sporozoite immunogenicity studies be performed using purified sporozoites whenever feasible.
Collapse
Affiliation(s)
- Zachary P Billman
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Annette M Seilie
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Sean C Murphy
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington, USA
| |
Collapse
|
25
|
Ishizuka AS, Lyke KE, DeZure A, Berry AA, Richie TL, Mendoza FH, Enama ME, Gordon IJ, Chang LJ, Sarwar UN, Zephir KL, Holman LA, James ER, Billingsley PF, Gunasekera A, Chakravarty S, Manoj A, Li M, Ruben AJ, Li T, Eappen AG, Stafford RE, K C N, Murshedkar T, DeCederfelt H, Plummer SH, Hendel CS, Novik L, Costner PJM, Saunders JG, Laurens MB, Plowe CV, Flynn B, Whalen WR, Todd JP, Noor J, Rao S, Sierra-Davidson K, Lynn GM, Epstein JE, Kemp MA, Fahle GA, Mikolajczak SA, Fishbaugher M, Sack BK, Kappe SHI, Davidson SA, Garver LS, Björkström NK, Nason MC, Graham BS, Roederer M, Sim BKL, Hoffman SL, Ledgerwood JE, Seder RA. Protection against malaria at 1 year and immune correlates following PfSPZ vaccination. Nat Med 2016; 22:614-23. [PMID: 27158907 PMCID: PMC11294733 DOI: 10.1038/nm.4110] [Citation(s) in RCA: 259] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/15/2016] [Indexed: 02/07/2023]
Abstract
An attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) vaccine, PfSPZ Vaccine, is highly protective against controlled human malaria infection (CHMI) 3 weeks after immunization, but the durability of protection is unknown. We assessed how vaccine dosage, regimen, and route of administration affected durable protection in malaria-naive adults. After four intravenous immunizations with 2.7 × 10(5) PfSPZ, 6/11 (55%) vaccinated subjects remained without parasitemia following CHMI 21 weeks after immunization. Five non-parasitemic subjects from this dosage group underwent repeat CHMI at 59 weeks, and none developed parasitemia. Although Pf-specific serum antibody levels correlated with protection up to 21-25 weeks after immunization, antibody levels waned substantially by 59 weeks. Pf-specific T cell responses also declined in blood by 59 weeks. To determine whether T cell responses in blood reflected responses in liver, we vaccinated nonhuman primates with PfSPZ Vaccine. Pf-specific interferon-γ-producing CD8 T cells were present at ∼100-fold higher frequencies in liver than in blood. Our findings suggest that PfSPZ Vaccine conferred durable protection to malaria through long-lived tissue-resident T cells and that administration of higher doses may further enhance protection.
Collapse
Affiliation(s)
- Andrew S Ishizuka
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Kirsten E Lyke
- Institute for Global Health, Center for Vaccine Development and Division of Malaria Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Adam DeZure
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Andrea A Berry
- Institute for Global Health, Center for Vaccine Development and Division of Malaria Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Floreliz H Mendoza
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Mary E Enama
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Ingelise J Gordon
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Lee-Jah Chang
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Uzma N Sarwar
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Kathryn L Zephir
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - LaSonji A Holman
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | | | | | | | | | | | - MingLin Li
- Sanaria Inc., Rockville, Maryland, USA
- Protein Potential, LLC, Rockville, Maryland, USA
| | | | - Tao Li
- Sanaria Inc., Rockville, Maryland, USA
| | | | - Richard E Stafford
- Sanaria Inc., Rockville, Maryland, USA
- Protein Potential, LLC, Rockville, Maryland, USA
| | - Natasha K C
- Sanaria Inc., Rockville, Maryland, USA
- Protein Potential, LLC, Rockville, Maryland, USA
| | | | - Hope DeCederfelt
- Pharmaceutical Development Section, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarah H Plummer
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Cynthia S Hendel
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Laura Novik
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Pamela J M Costner
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Jamie G Saunders
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Matthew B Laurens
- Institute for Global Health, Center for Vaccine Development and Division of Malaria Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Christopher V Plowe
- Institute for Global Health, Center for Vaccine Development and Division of Malaria Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Barbara Flynn
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - William R Whalen
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - J P Todd
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Jay Noor
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Srinivas Rao
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Kailan Sierra-Davidson
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Geoffrey M Lynn
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Judith E Epstein
- Naval Medical Research Center (NMRC), Malaria Department, Silver Spring, Maryland, USA
| | - Margaret A Kemp
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Gary A Fahle
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Brandon K Sack
- Center for Infectious Disease Research, Seattle, Washington, USA
| | - Stefan H I Kappe
- Center for Infectious Disease Research, Seattle, Washington, USA
| | - Silas A Davidson
- Entomology Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Lindsey S Garver
- Entomology Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Martha C Nason
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Barney S Graham
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Mario Roederer
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - B Kim Lee Sim
- Sanaria Inc., Rockville, Maryland, USA
- Protein Potential, LLC, Rockville, Maryland, USA
| | | | - Julie E Ledgerwood
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| | - Robert A Seder
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (NIH), Maryland, USA
| |
Collapse
|
26
|
De Niz M, Stanway RR, Wacker R, Keller D, Heussler VT. An ultrasensitive NanoLuc-based luminescence system for monitoring Plasmodium berghei throughout its life cycle. Malar J 2016; 15:232. [PMID: 27102897 PMCID: PMC4840902 DOI: 10.1186/s12936-016-1291-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/13/2016] [Indexed: 01/08/2023] Open
Abstract
Background Bioluminescence imaging is widely used for cell-based assays and animal imaging studies, both in biomedical research and drug development. Its main advantages include its high-throughput applicability, affordability, high sensitivity, operational simplicity, and quantitative outputs. In malaria research, bioluminescence has been used for drug discovery in vivo and in vitro, exploring host-pathogen interactions, and studying multiple aspects of Plasmodium biology. While the number of fluorescent proteins available for imaging has undergone a great expansion over the last two decades, enabling simultaneous visualization of multiple molecular and cellular events, expansion of available luciferases has lagged behind. The most widely used bioluminescent probe in malaria research is the Photinus pyralis firefly luciferase, followed by the more recently introduced Click-beetle and Renilla luciferases. Ultra-sensitive imaging of Plasmodium at low parasite densities has not been previously achieved. With the purpose of overcoming these challenges, a Plasmodium berghei line expressing the novel ultra-bright luciferase enzyme NanoLuc, called PbNLuc has been generated, and is presented in this work. Results NanoLuc shows at least 150 times brighter signal than firefly luciferase in vitro, allowing single parasite detection in mosquito, liver, and sexual and asexual blood stages. As a proof-of-concept, the PbNLuc parasites were used to image parasite development in the mosquito, liver and blood stages of infection, and to specifically explore parasite liver stage egress, and pre-patency period in vivo. Conclusions PbNLuc is a suitable parasite line for sensitive imaging of the entire Plasmodium life cycle. Its sensitivity makes it a promising line to be used as a reference for drug candidate testing, as well as the characterization of mutant parasites to explore the function of parasite proteins, host-parasite interactions, and the better understanding of Plasmodium biology. Since the substrate requirements of NanoLuc are different from those of firefly luciferase, dual bioluminescence imaging for the simultaneous characterization of two lines, or two separate biological processes, is possible, as demonstrated in this work.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland.
| | - Rebecca R Stanway
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland
| | - Rahel Wacker
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland
| | - Derya Keller
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland
| | - Volker T Heussler
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland
| |
Collapse
|
27
|
Taylor SL, Mason SKG, Glinton SL, Cobbold M, Dehghani H. Accounting for filter bandwidth improves the quantitative accuracy of bioluminescence tomography. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:096001. [PMID: 26325264 PMCID: PMC5996822 DOI: 10.1117/1.jbo.20.9.096001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 07/20/2015] [Indexed: 05/25/2023]
Abstract
Bioluminescence imaging is a noninvasive technique whereby surface weighted images of luminescent probes within animals are used to characterize cell count and function. Traditionally, data are collected over the entire emission spectrum of the source using no filters and are used to evaluate cell count/function over the entire spectrum. Alternatively, multispectral data over several wavelengths can be incorporated to perform tomographic reconstruction of source location and intensity. However, bandpass filters used for multispectral data acquisition have a specific bandwidth, which is ignored in the reconstruction. In this work, ignoring the bandwidth is shown to introduce a dependence of the recovered source intensity on the bandwidth of the filters. A method of accounting for the bandwidth of filters used during multispectral data acquisition is presented and its efficacy in increasing the quantitative accuracy of bioluminescence tomography is demonstrated through simulation and experiment. It is demonstrated that while using filters with a large bandwidth can dramatically decrease the data acquisition time, if not accounted for, errors of up to 200% in quantitative accuracy are introduced in two-dimensional planar imaging, even after normalization. For tomographic imaging, the use of this method to account for filter bandwidth dramatically improves the quantitative accuracy.
Collapse
Affiliation(s)
- Shelley L. Taylor
- University of Birmingham, PSIBS Doctoral Training Centre, Edgbaston, Birmingham B15 2TT, United Kingdom
- University of Birmingham, School of Computer Science, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Suzannah K. G. Mason
- University of Birmingham, PSIBS Doctoral Training Centre, Edgbaston, Birmingham B15 2TT, United Kingdom
- University of Birmingham, School of Computer Science, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Sophie L. Glinton
- University of Birmingham, PSIBS Doctoral Training Centre, Edgbaston, Birmingham B15 2TT, United Kingdom
- University of Birmingham, School of Computer Science, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Mark Cobbold
- University of Birmingham, College of Medical and Dental Sciences, School of Immunity and Infection, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Hamid Dehghani
- University of Birmingham, PSIBS Doctoral Training Centre, Edgbaston, Birmingham B15 2TT, United Kingdom
- University of Birmingham, School of Computer Science, Edgbaston, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
28
|
Li X, Kawamura A, Andrews CD, Miller JL, Wu D, Tsao T, Zhang M, Oren D, Padte NN, Porcelli SA, Wong CH, Kappe SHI, Ho DD, Tsuji M. Colocalization of a CD1d-Binding Glycolipid with a Radiation-Attenuated Sporozoite Vaccine in Lymph Node-Resident Dendritic Cells for a Robust Adjuvant Effect. THE JOURNAL OF IMMUNOLOGY 2015; 195:2710-21. [PMID: 26254338 DOI: 10.4049/jimmunol.1403017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 07/07/2015] [Indexed: 01/20/2023]
Abstract
A CD1d-binding glycolipid, α-Galactosylceramide (αGalCer), activates invariant NK T cells and acts as an adjuvant. We previously identified a fluorinated phenyl ring-modified αGalCer analog, 7DW8-5, displaying nearly 100-fold stronger CD1d binding affinity. In the current study, 7DW8-5 was found to exert a more potent adjuvant effect than αGalCer for a vaccine based on radiation-attenuated sporozoites of a rodent malaria parasite, Plasmodium yoelii, also referred to as irradiated P. yoelii sporozoites (IrPySpz). 7DW8-5 had a superb adjuvant effect only when the glycolipid and IrPySpz were conjointly administered i.m. Therefore, we evaluated the effect of distinctly different biodistribution patterns of αGalCer and 7DW8-5 on their respective adjuvant activities. Although both glycolipids induce a similar cytokine response in sera of mice injected i.v., after i.m. injection, αGalCer induces a systemic cytokine response, whereas 7DW8-5 is locally trapped by CD1d expressed by dendritic cells (DCs) in draining lymph nodes (dLNs). Moreover, the i.m. coadministration of 7DW8-5 with IrPySpz results in the recruitment of DCs to dLNs and the activation and maturation of DCs. These events cause the potent adjuvant effect of 7DW8-5, resulting in the enhancement of the CD8(+) T cell response induced by IrPySpz and, ultimately, improved protection against malaria. Our study is the first to show that the colocalization of a CD1d-binding invariant NK T cell-stimulatory glycolipid and a vaccine, like radiation-attenuated sporozoites, in dLN-resident DCs upon i.m. conjoint administration governs the potency of the adjuvant effect of the glycolipid.
Collapse
Affiliation(s)
- Xiangming Li
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016
| | - Akira Kawamura
- Department of Chemistry, Hunter College of The City University of New York, New York, NY 10065
| | - Chasity D Andrews
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016
| | | | - Douglass Wu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Tiffany Tsao
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016
| | - Min Zhang
- Department of Pathology, New York University, New York, NY 10016
| | - Deena Oren
- Structural Biology Resource Center, The Rockefeller University, New York, NY 10065
| | - Neal N Padte
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016
| | - Steven A Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461; and
| | - Chi-Huey Wong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037; Academia Sinica, Taipei 115-74, Taiwan, Republic of China
| | | | - David D Ho
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016;
| |
Collapse
|
29
|
de Koning-Ward TF, Gilson PR, Crabb BS. Advances in molecular genetic systems in malaria. Nat Rev Microbiol 2015; 13:373-87. [DOI: 10.1038/nrmicro3450] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
30
|
Siciliano G, Alano P. Enlightening the malaria parasite life cycle: bioluminescent Plasmodium in fundamental and applied research. Front Microbiol 2015; 6:391. [PMID: 26029172 PMCID: PMC4426725 DOI: 10.3389/fmicb.2015.00391] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/16/2015] [Indexed: 12/31/2022] Open
Abstract
The unicellular protozoan parasites of the genus Plasmodium impose on human health worldwide the enormous burden of malaria. The possibility to genetically modify several species of malaria parasites represented a major advance in the possibility to elucidate their biology and is now turning laboratory lines of transgenic Plasmodium into precious weapons to fight malaria. Amongst the various genetically modified plasmodia, transgenic parasite lines expressing bioluminescent reporters have been essential to unveil mechanisms of parasite gene expression and to develop in vivo imaging approaches in mouse malaria models. Mainly the human malaria parasite Plasmodium falciparum and the rodent parasite P. berghei have been engineered to express bioluminescent reporters in almost all the developmental stages of the parasite along its complex life cycle between the insect and the vertebrate hosts. Plasmodium lines expressing conventional and improved luciferase reporters are now gaining a central role to develop cell based assays in the much needed search of new antimalarial drugs and to open innovative approaches for both fundamental and applied research in malaria.
Collapse
Affiliation(s)
| | - Pietro Alano
- Dipartimento di Malattie Infettive, Parassitarie ed Immunomediate, Istituto Superiore di SanitàRome, Italy
| |
Collapse
|
31
|
De Niz M, Helm S, Horstmann S, Annoura T, del Portillo HA, Khan SM, Heussler VT. In vivo and in vitro characterization of a Plasmodium liver stage-specific promoter. PLoS One 2015; 10:e0123473. [PMID: 25874388 PMCID: PMC4398466 DOI: 10.1371/journal.pone.0123473] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 03/03/2015] [Indexed: 12/28/2022] Open
Abstract
Little is known about stage-specific gene regulation in Plasmodium parasites, in particular the liver stage of development. We have previously described in the Plasmodium berghei rodent model, a liver stage-specific (lisp2) gene promoter region, in vitro. Using a dual luminescence system, we now confirm the stage specificity of this promoter region also in vivo. Furthermore, by substitution and deletion analyses we have extended our in vitro characterization of important elements within the promoter region. Importantly, the dual luminescence system allows analyzing promoter constructs avoiding mouse-consuming cloning procedures of transgenic parasites. This makes extensive mutation and deletion studies a reasonable approach also in the malaria mouse model. Stage-specific expression constructs and parasite lines are extremely valuable tools for research on Plasmodium liver stage biology. Such reporter lines offer a promising opportunity for assessment of liver stage drugs, characterization of genetically attenuated parasites and liver stage-specific vaccines both in vivo and in vitro, and may be key for the generation of inducible systems.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- * E-mail:
| | - Susanne Helm
- Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Sebastian Horstmann
- Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Takeshi Annoura
- Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Department of Parasitology, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| | - Hernando A. del Portillo
- Barcelona Centre for International Health Research (CRESIB), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Shahid M. Khan
- Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
32
|
Sahu T, Lambert L, Herrod J, Conteh S, Orr-Gonzalez S, Carter D, Duffy PE. Chloroquine neither eliminates liver stage parasites nor delays their development in a murine Chemoprophylaxis Vaccination model. Front Microbiol 2015; 6:283. [PMID: 25914686 PMCID: PMC4391028 DOI: 10.3389/fmicb.2015.00283] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 03/22/2015] [Indexed: 11/13/2022] Open
Abstract
Chemoprophylaxis Vaccination (CVac) confers long lasting sterile protection against homologous parasite strains in humans, and involves inoculation of infectious sporozoites (SPZ) under drug cover. CVac using the drug chloroquine (CQ) induces pre-erythrocytic immunity in humans that includes antibody to SPZ and T-cell responses to liver stage (LS) parasites. The mechanism by which CVac with CQ induces strong protective immunity is not understood as untreated infections do not confer protection. CQ kills blood stage parasites, but its effect on LS parasites is poorly studied. Here we hypothesized that CQ may prolong or perturb LS development of Plasmodium, as a potential explanation for enhanced pre-erythrocytic immune responses. Balb/c mice with or without CQ prophylaxis were infected with sporozoite forms of a luciferase-expressing rodent parasite, Plasmodium yoelii-Luc (Py-Luc). Mice that received primaquine, a drug that kills LS parasites, served as a positive control of drug effect. Parasite burden in liver was measured both by bioluminescence and by qRT-PCR quantification of parasite transcript. Time to appearance of parasites in the blood was monitored by microscopic analysis of Giemsa-stained thick and thin blood smears. The parasite load in livers of CQ-treated and untreated mice did not significantly differ at any of the time points studied. Parasites appeared in the blood smears of both CQ-treated and untreated mice 3 days after infection. Taken together, our findings confirm that CQ neither eliminates LS parasites nor delays their development. Further investigations into the mechanism of CQ-induced protection after CVac are required, and may give insights relevant to drug and vaccine development.
Collapse
Affiliation(s)
- Tejram Sahu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville MD, USA
| | - Lynn Lambert
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville MD, USA
| | - Jessica Herrod
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville MD, USA
| | - Solomon Conteh
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville MD, USA
| | - Sachy Orr-Gonzalez
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville MD, USA
| | - Dariyen Carter
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville MD, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville MD, USA
| |
Collapse
|
33
|
Matz JM, Kooij TWA. Towards genome-wide experimental genetics in the in vivo malaria model parasite Plasmodium berghei. Pathog Glob Health 2015; 109:46-60. [PMID: 25789828 DOI: 10.1179/2047773215y.0000000006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Plasmodium berghei was identified as a parasite of thicket rats (Grammomys dolichurus) and Anopheles dureni mosquitoes in African highland forests. Successful adaptation to a range of rodent and mosquito species established P. berghei as a malaria model parasite. The introduction of stable transfection technology, permitted classical reverse genetics strategies and thus systematic functional profiling of the gene repertoire. In the past 10 years following the publication of the P. berghei genome sequence, many new tools for experimental genetics approaches have been developed and existing ones have been improved. The infection of mice is the principal limitation towards a genome-wide repository of mutant parasite lines. In the past few years, there have been some promising and most welcome developments that allow rapid selection and isolation of recombinant parasites while simultaneously minimising animal usage. Here, we provide an overview of all the currently available tools and methods.
Collapse
|
34
|
Murray SA, Mohar I, Miller JL, Brempelis KJ, Vaughan AM, Kappe SHI, Crispe IN. CD40 is required for protective immunity against liver stage Plasmodium infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:2268-79. [PMID: 25646303 DOI: 10.4049/jimmunol.1401724] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The costimulatory molecule CD40 enhances immunity through several distinct roles in T cell activation and T cell interaction with other immune cells. In a mouse model of immunity to liver stage Plasmodium infection, CD40 was critical for the full maturation of liver dendritic cells, accumulation of CD8(+) T cells in the liver, and protective immunity induced by immunization with the Plasmodium yoelii fabb/f(-) genetically attenuated parasite. Using mixed adoptive transfers of polyclonal wild-type and CD40-deficient CD8(+) T cells into wild-type and CD40-deficient hosts, we evaluated the contributions to CD8(+) T cell immunity of CD40 expressed on host tissues including APC, compared with CD40 expressed on the CD8(+) T cells themselves. Most of the effects of CD40 could be accounted for by expression in the T cells' environment, including the accumulation of large numbers of CD8(+) T cells in the livers of immunized mice. Thus, protective immunity generated during immunization with fabb/f(-) was largely dependent on effective APC licensing via CD40 signaling.
Collapse
Affiliation(s)
- Sara A Murray
- Department of Global Health, University of Washington, Seattle, WA 98195
| | - Isaac Mohar
- Department of Pathology, University of Washington, Seattle, WA 98195; and
| | | | | | | | - Stefan H I Kappe
- Department of Global Health, University of Washington, Seattle, WA 98195; Seattle Biomedical Research Institute, Seattle, WA 98109
| | - Ian N Crispe
- Department of Pathology, University of Washington, Seattle, WA 98195; and
| |
Collapse
|
35
|
Sack BK, Miller JL, Vaughan AM, Kappe SHI. Measurement of Antibody-Mediated Reduction of Plasmodium yoelii Liver Burden by Bioluminescent Imaging. Methods Mol Biol 2015; 1325:69-80. [PMID: 26450380 PMCID: PMC8441651 DOI: 10.1007/978-1-4939-2815-6_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Antibodies against the infectious sporozoite stage of malaria have been shown to be effective in preventing infection of the liver and in mitigating the ensuing blood stage. However, only a handful of antibody targets have been vetted and shown to be successful in mediating in vivo protection. Even more limited are the means with which to measure how effectively antibodies can reduce the number of parasites establishing infection in the liver. Traditionally, only qPCR of infected mouse livers could accurately measure liver parasite burden. However, this procedure requires sacrifice of the animal and precludes monitoring of the ensuing blood stage infection. Herein we describe a method of accurately assessing antibody-mediated reduction of parasite liver burden by combining passive or active immunization of mice and mosquito bite challenge with luciferase-expressing transgenic P. yoelii parasites. This method is rapid, reliable and allows for observation of blood stage disease in the same animal. This model will prove integral in screening the efficacy of novel antibody targets as the search for a more effective malaria vaccine continues.
Collapse
Affiliation(s)
- Brandon K Sack
- Center for Infectious Disease Research, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA.
| | - Jessica L Miller
- Center for Infectious Disease Research, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Ashley M Vaughan
- Center for Infectious Disease Research, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Stefan H I Kappe
- Center for Infectious Disease Research, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| |
Collapse
|
36
|
Matsuoka H, Tomita H, Hattori R, Arai M, Hirai M. Visualization of Malaria Parasites in the Skin Using the Luciferase Transgenic Parasite, Plasmodium berghei. Trop Med Health 2014; 43:53-61. [PMID: 25859153 PMCID: PMC4361344 DOI: 10.2149/tmh.2014-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/13/2014] [Indexed: 01/25/2023] Open
Abstract
We produced a transgenic rodent malaria parasite (Plasmodium berghei) that contained the luciferase gene under a promoter region of elongation factor-1α. These transgenic (TG) parasites expressed luciferase in all stages of their life cycle, as previously reported. However, we were the first to succeed in observing sporozoites as a mass in mouse skin following their deposition by the probing of infective mosquitoes. Our transgenic parasites may have emitted stronger bioluminescence than previous TG parasites. The estimated number of injected sporozoites by mosquitoes was between 34 and 775 (median 80). Since luciferase activity diminished immediately after the death of the parasites, luciferase activity could be an indicator of the existence of live parasites. Our results indicated that sporozoites survived at the probed site for more than 42 hours. We also detected sporozoites in the liver within 15 min of the intravenous injection. Bioluminescence was not observed in the lung, kidney or spleen. We confirmed the observation that the liver was the first organ in which malaria parasites entered and increased in number.
Collapse
Affiliation(s)
- Hiroyuki Matsuoka
- Division of Medical Zoology, Jichi Medical University , 3311-1 Yakushiji, Shimotsuke-shi 329-0498, Japan
| | - Hiroyuki Tomita
- Division of Medical Zoology, Jichi Medical University , 3311-1 Yakushiji, Shimotsuke-shi 329-0498, Japan
| | - Ryuta Hattori
- Division of Medical Zoology, Jichi Medical University , 3311-1 Yakushiji, Shimotsuke-shi 329-0498, Japan
| | - Meiji Arai
- Division of Medical Zoology, Jichi Medical University , 3311-1 Yakushiji, Shimotsuke-shi 329-0498, Japan ; Department of International Medical Zoology, Graduate School of Medicine, Kagawa University , Miki-cho 761-0793, Japan
| | - Makoto Hirai
- Division of Medical Zoology, Jichi Medical University , 3311-1 Yakushiji, Shimotsuke-shi 329-0498, Japan ; Department of Parasitology, School of Medicine, Juntendo University , Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
37
|
Susceptibility to Plasmodium yoelii preerythrocytic infection in BALB/c substrains is determined at the point of hepatocyte invasion. Infect Immun 2014; 83:39-47. [PMID: 25312960 DOI: 10.1128/iai.02230-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
After transmission by Anopheles mosquitoes, Plasmodium sporozoites travel to the liver, infect hepatocytes, and rapidly develop as intrahepatocytic liver stages (LS). Rodent models of malaria exhibit large differences in the magnitude of liver infection, both between parasite species and between strains of mice. This has been mainly attributed to differences in innate immune responses and parasite infectivity. Here, we report that BALB/cByJ mice are more susceptible to Plasmodium yoelii preerythrocytic infection than BALB/cJ mice. This difference occurs at the level of early hepatocyte infection, but expression levels of reported host factors that are involved in infection do not correlate with susceptibility. Interestingly, BALB/cByJ hepatocytes are more frequently polyploid; thus, their susceptibility converges on the previously observed preference of sporozoites to infect polyploid hepatocytes. Gene expression analysis demonstrates hepatocyte-specific differences in mRNA abundance for numerous genes between BALB/cByJ and BALB/cJ mice, some of which encode hepatocyte surface molecules. These data suggest that a yet-unknown receptor for sporozoite infection, present at elevated levels on BALB/cByJ hepatocytes and also polyploid hepatocytes, might facilitate Plasmodium liver infection.
Collapse
|
38
|
Immunization of mice with live-attenuated late liver stage-arresting Plasmodium yoelii parasites generates protective antibody responses to preerythrocytic stages of malaria. Infect Immun 2014; 82:5143-53. [PMID: 25267837 DOI: 10.1128/iai.02320-14] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Understanding protective immunity to malaria is essential for the design of an effective vaccine to prevent the large number of infections and deaths caused by this parasitic disease. To date, whole-parasite immunization with attenuated parasites is the most effective method to confer sterile protection against malaria infection in clinical trials. Mouse model studies have highlighted the essential role that CD8(+) T cells play in protection against preerythrocytic stages of malaria; however, there is mounting evidence that antibodies are also important in these stages. Here, we show that experimental immunization of mice with Plasmodium yoelii fabb/f(-) (Pyfabb/f(-)), a genetically attenuated rodent malaria parasite that arrests late in the liver stage, induced functional antibodies that inhibited hepatocyte invasion in vitro and reduced liver-stage burden in vivo. These antibodies were sufficient to induce sterile protection from challenge by P. yoelii sporozoites in the absence of T cells in 50% of mice when sporozoites were administered by mosquito bite but not when they were administered by intravenous injection. Moreover, among mice challenged by mosquito bite, a higher proportion of BALB/c mice than C57BL/6 mice developed sterile protection (62.5% and 37.5%, respectively). Analysis of the antibody isotypes induced by immunization with Pyfabb/f(-) showed that, overall, BALB/c mice developed an IgG1-biased response, whereas C57BL/6 mice developed an IgG2b/c-biased response. Our data demonstrate for the first time that antibodies induced by experimental immunization of mice with a genetically attenuated rodent parasite play a protective role during the preerythrocytic stages of malaria. Furthermore, they highlight the importance of considering both the route of challenge and the genetic background of the mouse strains used when interpreting vaccine efficacy studies in animal models of malaria infection.
Collapse
|
39
|
Chen L, Keitany GJ, Peng X, Gibson C, Mohar I, Vignali M, Crispe IN, Huang F, Wang R. Identification of pre-erythrocytic malaria antigens that target hepatocytes for killing in vivo and contribute to protection elicited by whole-parasite vaccination. PLoS One 2014; 9:e102225. [PMID: 25025375 PMCID: PMC4099202 DOI: 10.1371/journal.pone.0102225] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 06/17/2014] [Indexed: 11/19/2022] Open
Abstract
Pre-erythrocytic malaria vaccines, including those based on whole-parasite approaches, have shown protective efficacy in animal and human studies. However few pre-erythocytic antigens other than the immunodominant circumsporozoite protein (CSP) have been studied in depth with the goal of developing potent subunit malaria vaccines that are suited for use in endemic areas. Here we describe a novel technique to identify pre-erythrocytic malaria antigens that contribute to protection elicited by whole-parasite vaccination in the mouse model. Our approach combines immunization with genetically attenuated parasites and challenge with DNA plasmids encoding for potential protective pre-erythrocytic malaria antigens as luciferase fusions by hydrodynamic tail vein injection. After optimizing the technique, we first showed that immunization with Pyfabb/f−, a P. yoelii genetically attenuated parasite, induces killing of CSP-presenting hepatocytes. Depletion of CD8+ but not CD4+ T cells diminished the killing of CSP-expressing hepatocytes, indicating that killing is CD8+ T cell-dependent. Finally we showed that the use of heterologous prime/boost immunization strategies that use genetically attenuated parasites and DNA vaccines enabled the characterization of a novel pre-erythrocytic antigen, Tmp21, as a contributor to Pyfabb/f− induced protection. This technique will be valuable for identification of potentially protective liver stage antigens and has the potential to contribute to the understanding of immunity elicited by whole parasite vaccination, as well as the development of effective subunit malaria vaccines.
Collapse
Affiliation(s)
- Lin Chen
- Department of Pathogenic Biology, Third Military Medical University, Chongqing, China
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Gladys J. Keitany
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Xiaohong Peng
- Department of Pathogenic Biology, Third Military Medical University, Chongqing, China
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Claire Gibson
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Isaac Mohar
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Marissa Vignali
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Ian N. Crispe
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Fusheng Huang
- Department of Pathogenic Biology, Third Military Medical University, Chongqing, China
- * E-mail: (FH); (RW)
| | - Ruobing Wang
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- * E-mail: (FH); (RW)
| |
Collapse
|
40
|
Peng X, Keitany GJ, Vignali M, Chen L, Gibson C, Choi K, Huang F, Wang R. Artesunate versus chloroquine infection-treatment-vaccination defines stage-specific immune responses associated with prolonged sterile protection against both pre-erythrocytic and erythrocytic Plasmodium yoelii infection. THE JOURNAL OF IMMUNOLOGY 2014; 193:1268-77. [PMID: 24958899 DOI: 10.4049/jimmunol.1400296] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sterile protection against malaria infection can be achieved through vaccination of mice and humans with whole Plasmodium spp. parasites. One such method, known as infection-treatment-vaccination (ITV), involves immunization with wild type sporozoites (spz) under drug coverage. In this work, we used the different effects of antimalarial drugs chloroquine (CQ) and artesunate (AS) on blood stage (BS) parasites to dissect the stage-specific immune responses in mice immunized with Plasmodium yoelii spz under either drug, as well as their ability to protect mice against challenge with spz or infected RBCs (iRBCs). Whereas CQ-ITV induced sterile protection against challenge with both spz and iRBCs, AS-ITV only induced sterile protection against spz challenge. Importantly, AS-ITV delayed the onset of BS infection, indicating that both regimens induced cross-stage immunity. Moreover, both CQ- and AS-ITV induced CD8(+) T cells in the liver that eliminated malaria-infected hepatocytes in vitro, as well as Abs that recognized pre-erythrocytic parasites. Sera from both groups of mice inhibited spz invasion of hepatocytes in vitro, but only CQ-ITV induced high levels of anti-BS Abs. Finally, passive transfer of sera from CQ-ITV-treated mice delayed the onset of erythrocytic infection in the majority of mice challenged with P. yoelii iRBCs. Besides constituting the first characterization, to our knowledge, of AS-ITV as a vaccination strategy, our data show that ITV strategies that lead to subtle differences in the persistence of parasites in the blood enable the characterization of the resulting immune responses, which will contribute to future research in vaccine design and malaria interventions.
Collapse
Affiliation(s)
- Xiaohong Peng
- Department of Pathogenic Biology, Third Military Medical University, Chongqing 400038, China; Seattle Biomedical Research Institute, Seattle, WA 98109; and
| | | | - Marissa Vignali
- Seattle Biomedical Research Institute, Seattle, WA 98109; and
| | - Lin Chen
- Department of Pathogenic Biology, Third Military Medical University, Chongqing 400038, China; Seattle Biomedical Research Institute, Seattle, WA 98109; and
| | - Claire Gibson
- Seattle Biomedical Research Institute, Seattle, WA 98109; and
| | - Kimberly Choi
- Seattle Biomedical Research Institute, Seattle, WA 98109; and
| | - Fusheng Huang
- Department of Pathogenic Biology, Third Military Medical University, Chongqing 400038, China
| | - Ruobing Wang
- Seattle Biomedical Research Institute, Seattle, WA 98109; and Department of Global Health, University of Washington, Seattle, WA 98125
| |
Collapse
|
41
|
Li Q, O'Neil M, Xie L, Caridha D, Zeng Q, Zhang J, Pybus B, Hickman M, Melendez V. Assessment of the prophylactic activity and pharmacokinetic profile of oral tafenoquine compared to primaquine for inhibition of liver stage malaria infections. Malar J 2014; 13:141. [PMID: 24731238 PMCID: PMC3989846 DOI: 10.1186/1475-2875-13-141] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/02/2014] [Indexed: 11/10/2022] Open
Abstract
Background As anti-malarial drug resistance escalates, new safe and effective medications are necessary to prevent and treat malaria infections. The US Army is developing tafenoquine (TQ), an analogue of primaquine (PQ), which is expected to be more effective in preventing malaria in deployed military personnel. Methods To compare the prophylactic efficacy of TQ and PQ, a transgenic Plasmodium berghei parasite expressing the bioluminescent reporter protein luciferase was utilized to visualize and quantify parasite development in C57BL/6 albino mice treated with PQ and TQ in single or multiple regimens using a real-time in vivo imaging system (IVIS). As an additional endpoint, blood stage parasitaemia was monitored by flow cytometry. Comparative pharmacokinetic (PK) and liver distribution studies of oral and intravenous PQ and TQ were also performed. Results Mice treated orally with three doses of TQ at 5 mg/kg three doses of PQ at 25 mg/kg demonstrated no bioluminescence liver signal and no blood stage parasitaemia was observed suggesting both drugs showed 100% causal activity at the doses tested. Single dose oral treatment with 5 mg TQ or 25 mg of PQ, however, yielded different results as only TQ treatment resulted in causal prophylaxis in P. berghei sporozoite-infected mice. TQ is highly effective for causal prophylaxis in mice at a minimal curative single oral dose of 5 mg/kg, which is a five-fold improvement in potency versus PQ. PK studies of the two drugs administered orally to mice showed that the absolute bioavailability of oral TQ was 3.5-fold higher than PQ, and the AUC of oral TQ was 94-fold higher than oral PQ. The elimination half-life of oral TQ in mice was 28 times longer than PQ, and the liver tissue distribution of TQ revealed an AUC that was 188-fold higher than PQ. Conclusions The increased drug exposure levels and longer exposure time of oral TQ in the plasma and livers of mice highlight the lead quality attributes that explain the much improved efficacy of TQ when compared to PQ.
Collapse
Affiliation(s)
- Qigui Li
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Miller J, Sack B, Baldwin M, Vaughan A, Kappe S. Interferon-Mediated Innate Immune Responses against Malaria Parasite Liver Stages. Cell Rep 2014; 7:436-447. [DOI: 10.1016/j.celrep.2014.03.018] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 02/07/2014] [Accepted: 03/11/2014] [Indexed: 01/25/2023] Open
|
43
|
Zuzarte-Luis V, Sales-Dias J, Mota MM. Simple, sensitive and quantitative bioluminescence assay for determination of malaria pre-patent period. Malar J 2014; 13:15. [PMID: 24400642 PMCID: PMC3893453 DOI: 10.1186/1475-2875-13-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 12/31/2013] [Indexed: 12/22/2022] Open
Abstract
Background The first phase of malaria infection occurs in the liver and is clinically silent. Inside hepatocytes each Plasmodium sporozoite replicate into thousands of erythrocyte-infectious merozoites that when released into the blood stream result in clinical symptoms of the disease. The time between sporozoite inoculation and the appearance of parasites in the blood is defined as the pre-patent period, which is classically analysed by time-consuming and labor-intensive techniques, such as microscopy and PCR. Methods Luciferase-expressing Plasmodium berghei parasites were used to measure pre-patent period of malaria infection in rodents using a bioluminescence assay that requires only one microliter of blood collected from the tail-vein. The accuracy and sensitivity of this new method was compared with conventional microscopy and PCR based techniques, and its capacity to measure the impact of anti-malarial interventions against the liver evaluated. Results The described method is very sensitive allowing the detection of parasites during the first cycles of blood stage replication. It accurately translates differences in liver load due to inoculation of different sporozoite doses as well as a result of treatment with different primaquine regimens. Conclusions A novel, simple, fast, and sensitive method to measure pre-patent period of malaria infection in rodents is described here. The sensitivity and accuracy of this new method is comparable to standard PCR and microscopy-based techniques, respectively.
Collapse
Affiliation(s)
| | | | - Maria M Mota
- Instituto de Medicina Molecular, Faculdade de Medicina Universidade de Lisboa, Lisboa 1649-028, Portugal.
| |
Collapse
|
44
|
Model for in vivo assessment of humoral protection against malaria sporozoite challenge by passive transfer of monoclonal antibodies and immune serum. Infect Immun 2013; 82:808-17. [PMID: 24478094 DOI: 10.1128/iai.01249-13] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Evidence from clinical trials of malaria vaccine candidates suggests that both cell-mediated and humoral immunity to pre-erythrocytic parasite stages can provide protection against infection. Novel pre-erythrocytic antibody (Ab) targets could be key to improving vaccine formulations, which are currently based on targeting antigens such as the circumsporozoite protein (CSP). However, methods to assess the effects of sporozoite-specific Abs on pre-erythrocytic infection in vivo remain underdeveloped. Here, we combined passive transfer of monoclonal Abs (MAbs) or immune serum with a luciferase-expressing Plasmodium yoelii sporozoite challenge to assess Ab-mediated inhibition of liver infection in mice. Passive transfer of a P. yoelii CSP MAb showed inhibition of liver infection when mice were challenged with sporozoites either intravenously or by infectious mosquito bite. However, inhibition was most potent for the mosquito bite challenge, leading to a more significant reduction of liver-stage burden and even a lack of progression to blood-stage parasitemia. This suggests that Abs provide effective protection against a natural infection. Inhibition of liver infection was also achieved by passive transfer of immune serum from whole-parasite-immunized mice. Furthermore, we demonstrated that passive transfer of a MAb against P. falciparum CSP inhibited liver-stage infection in a humanized mouse/P. falciparum challenge model. Together, these models constitute unique and sensitive in vivo methods to assess serum-transferable protection against Plasmodium sporozoite challenge.
Collapse
|
45
|
Holland AD, Rückerl F, Dragavon JM, Rekiki A, Tinevez JY, Tournebize R, Shorte SL. In vitro characterization of Fluorescence by Unbound Excitation from Luminescence: broadening the scope of energy transfer. Methods 2013; 66:353-61. [PMID: 24045025 DOI: 10.1016/j.ymeth.2013.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 08/20/2013] [Accepted: 09/02/2013] [Indexed: 01/10/2023] Open
Abstract
Energy transfer mechanisms represent the basis for an array of valuable tools to infer interactions in vitro and in vivo, enhance detection or resolve interspecies distances such as with resonance. Based upon our own previously published studies and new results shown here we present a novel framework describing for the first time a model giving a view of the biophysical relationship between Fluorescence by Unbound Excitation from Luminescence (FUEL), a conventional radiative excitation-emission process, and bioluminescence resonance energy transfer. We show here that in homogeneous solutions and in fluorophore-targeted bacteria, FUEL is the dominant mechanism responsible for the production of red-shifted photons. The minor resonance contribution was ascertained by comparing the intensity of the experimental signal to its theoretical resonance counterpart. Distinctive features of the in vitro FUEL signal include a macroscopic depth dependency, a lack of enhancement upon targeting at a constant fluorophore concentration cf and a non-square dependency on cf. Significantly, FUEL is an important, so far overlooked, component of all resonance phenomena which should guide the design of appropriate controls when elucidating interactions. Last, our results highlight the potential for FUEL as a means to enhance in vivo and in vitro detection through complex media while alleviating the need for targeting.
Collapse
Affiliation(s)
- Alexandra D Holland
- Plate-Forme d'Imagerie Dynamique, Imagopole, Institut Pasteur, 25-28 Rue du Dr. Roux, 75724 Paris cedex 15, France
| | - Florian Rückerl
- Plate-Forme d'Imagerie Dynamique, Imagopole, Institut Pasteur, 25-28 Rue du Dr. Roux, 75724 Paris cedex 15, France
| | - Joseph M Dragavon
- Plate-Forme d'Imagerie Dynamique, Imagopole, Institut Pasteur, 25-28 Rue du Dr. Roux, 75724 Paris cedex 15, France
| | - Abdessalem Rekiki
- Plate-Forme d'Imagerie Dynamique, Imagopole, Institut Pasteur, 25-28 Rue du Dr. Roux, 75724 Paris cedex 15, France
| | - Jean-Yves Tinevez
- Plate-Forme d'Imagerie Dynamique, Imagopole, Institut Pasteur, 25-28 Rue du Dr. Roux, 75724 Paris cedex 15, France
| | - Régis Tournebize
- Plate-Forme d'Imagerie Dynamique, Imagopole, Institut Pasteur, 25-28 Rue du Dr. Roux, 75724 Paris cedex 15, France; Unité INSERM U786, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris cedex 15, France.
| | - Spencer L Shorte
- Plate-Forme d'Imagerie Dynamique, Imagopole, Institut Pasteur, 25-28 Rue du Dr. Roux, 75724 Paris cedex 15, France.
| |
Collapse
|
46
|
In vivo imaging in NHP models of malaria: challenges, progress and outlooks. Parasitol Int 2013; 63:206-15. [PMID: 24042056 PMCID: PMC7108422 DOI: 10.1016/j.parint.2013.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 08/30/2013] [Accepted: 09/06/2013] [Indexed: 12/22/2022]
Abstract
Animal models of malaria, mainly mice, have made a large contribution to our knowledge of host-pathogen interactions and immune responses, and to drug and vaccine design. Non-human primate (NHP) models for malaria are admittedly under-used, although they are probably closer models than mice for human malaria; in particular, NHP models allow the use of human pathogens (Plasmodium falciparum, Plasmodium vivax, Plasmodium malariae and Plasmodium knowlesi). NHPs, whether natural hosts or experimentally challenged with a simian Plasmodium, can also serve as robust pre-clinical models. Some simian parasites are closely related to a human counterpart, with which they may share a common ancestor, and display similar major features with the human infection and pathology. NHP models allow longitudinal studies, from the early events following sporozoite inoculation to the later events, including analysis of organs and tissues, particularly liver, spleen, brain and bone marrow. NHP models have one other significant advantage over mouse models: NHPs are our closest relatives and thus their biology is very similar to ours. Recently developed in vivo imaging tools have provided insight into malaria parasite infection and disease in mouse models. One advantage of these tools is that they limit the need for invasive procedures, such as tissue biopsies. Many such technologies are now available for NHP studies and provide new opportunities for elucidating host/parasite interactions. The aim of this review is to bring the malaria community up to date on what is currently possible and what soon will be, in terms of in vivo imaging in NHP models of malaria, to consider the pros and the cons of the various techniques, and to identify challenges.
Collapse
|
47
|
Teranishi H, Schwartz RE, March S, Galstian A, Gural N, Shan J, Prabhu M, Mota MM, Bhatia SN. [Infection of Schwann cells along corneal stromal nerve fiber in experimental herpes simplex keratitis]. Stem Cell Reports 1975; 4:348-59. [PMID: 25660406 PMCID: PMC4375936 DOI: 10.1016/j.stemcr.2015.01.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 01/04/2015] [Accepted: 01/05/2015] [Indexed: 02/06/2023] Open
Abstract
Malaria eradication is a major goal in public health but is challenged by relapsing malaria species, expanding drug resistance, and the influence of host genetics on antimalarial drug efficacy. To overcome these hurdles, it is imperative to establish in vitro assays of liver-stage malaria for drug testing. Induced pluripotent stem cells (iPSC) potentially allow the assessment of donor-specific drug responses, and iPSC-derived hepatocyte-like cells (iHLCs) can facilitate the study of host genetics on host-pathogen interactions and the discovery of novel targets for antimalarial drug development. We establish in vitro liver-stage malaria infections in iHLCs using P. berghei, P. yoelii, P. falciparum, and P. vivax and show that differentiating cells acquire permissiveness to malaria infection at the hepatoblast stage. We also characterize antimalarial drug metabolism capabilities of iHLCs using prototypical antimalarial drugs and demonstrate that chemical maturation of iHLCs can improve their potential for antimalarial drug testing applications. iPSC-derived hepatocyte-like cells (iHLCs) can host liver-stage malaria in vitro iHLCs become permissive to Plasmodium infection at the hepatoblast stage Plasmodium-infected iHLCs are sensitive to atovaquone but not primaquine Small molecule-mediated maturation of iHLCs confers primaquine sensitivity
Collapse
Affiliation(s)
| | - Robert E Schwartz
- Health Sciences and Technology/Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Sandra March
- Health Sciences and Technology/Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute, Cambridge, MA 02139, USA
| | - Ani Galstian
- Health Sciences and Technology/Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute, Cambridge, MA 02139, USA
| | - Nil Gural
- Health Sciences and Technology/Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jing Shan
- Health Sciences and Technology/Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mythili Prabhu
- Health Sciences and Technology/Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Maria M Mota
- Unidade de Malária, Instituto de Medicina Molecular, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Howard Hughes Medical Institute, Koch Institute, and Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sangeeta N Bhatia
- Health Sciences and Technology/Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Koch Institute, and Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute, Cambridge, MA 02139, USA.
| |
Collapse
|