Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Sci Adv ; 10(17): eadm9281, 2024 Apr 26.
Article En | MEDLINE | ID: mdl-38657074

Critical aspects of physiology and cell function exhibit self-sustained ~24-hour variations termed circadian rhythms. In the liver, circadian rhythms play fundamental roles in maintaining organ homeostasis. Here, we established and characterized an in vitro liver experimental system in which primary human hepatocytes display self-sustained oscillations. By generating gene expression profiles of these hepatocytes over time, we demonstrated that their transcriptional state is dynamic across 24 hours and identified a set of cycling genes with functions related to inflammation, drug metabolism, and energy homeostasis. We designed and tested a treatment protocol to minimize atorvastatin- and acetaminophen-induced hepatotoxicity. Last, we documented circadian-dependent induction of pro-inflammatory cytokines when triggered by LPS, IFN-ß, or Plasmodium infection in human hepatocytes. Collectively, our findings emphasize that the phase of the circadian cycle has a robust impact on the efficacy and toxicity of drugs, and we provide a test bed to study the timing and magnitude of inflammatory responses over the course of infection in human liver.


Circadian Rhythm , Hepatocytes , Inflammation , Liver , Humans , Hepatocytes/metabolism , Hepatocytes/drug effects , Inflammation/metabolism , Liver/metabolism , Acetaminophen/pharmacology , Atorvastatin/pharmacology , Cytokines/metabolism , Inactivation, Metabolic , Lipopolysaccharides/pharmacology , Gene Expression Profiling , Gene Expression Regulation , Cells, Cultured
2.
Aust J Rural Health ; 31(3): 452-463, 2023 Jun.
Article En | MEDLINE | ID: mdl-36583508

OBJECTIVE: The purpose of this study was to explore the benefits of a partnership between a university dental school and a Community Controlled Health Service, specifically in relation to improving the oral health of an underserved rural Indigenous community. We sought community opinions on health and social outcomes arising from the service provided by the dental student clinical outplacement. SETTING: In Dalby, Queensland. PARTICIPANTS: In total, 38 participants in five focus groups were representative of local Indigenous community Elders, community health support group members and management and staff. DESIGN: A descriptive qualitative study employing semi-structured audio-recorded focus group discussions conducted with purposefully selected Indigenous community groups to explore participant views and experience of the partnership model. Qualitative data were analysed using thematic content analysis. RESULTS: The Indigenous community representatives expressed positive benefit in both their general and oral health awareness, in improved access to dental care provided in their own safe space, while they were pleased to assist with students' learning. They viewed the partnership as mutually beneficial. Suggestions for enhancement of the oral health service were also offered. CONCLUSION: This partnership between a university and a Community Controlled Health Service provides sustainable positive social and health benefits for the targeted Indigenous community and for the wider local population, while simultaneously providing enhanced educational benefits for students on clinical outplacement. Translation and uptake of this successful model of care would benefit both underserved communities and dental and other health care professional educators worldwide.


Health Services, Indigenous , Oral Health , Humans , Aged , Queensland , Community Health Services , Focus Groups , Qualitative Research
3.
Cell Host Microbe ; 30(7): 1048-1060.e5, 2022 07 13.
Article En | MEDLINE | ID: mdl-35443155

Malaria-causing Plasmodium vivax parasites can linger in the human liver for weeks to years and reactivate to cause recurrent blood-stage infection. Although they are an important target for malaria eradication, little is known about the molecular features of replicative and non-replicative intracellular liver-stage parasites and their host cell dependence. Here, we leverage a bioengineered human microliver platform to culture patient-derived P. vivax parasites for transcriptional profiling. Coupling enrichment strategies with bulk and single-cell analyses, we capture both parasite and host transcripts in individual hepatocytes throughout the course of infection. We define host- and state-dependent transcriptional signatures and identify unappreciated populations of replicative and non-replicative parasites that share features with sexual transmissive forms. We find that infection suppresses the transcription of key hepatocyte function genes and elicits an anti-parasite innate immune response. Our work provides a foundation for understanding host-parasite interactions and reveals insights into the biology of P. vivax dormancy and transmission.


Malaria, Vivax , Malaria , Hepatocytes/parasitology , Humans , Liver/parasitology , Malaria/parasitology , Malaria, Vivax/parasitology , Plasmodium vivax/genetics
4.
Nature ; 595(7866): 289-294, 2021 07.
Article En | MEDLINE | ID: mdl-34194041

The global decline in malaria has stalled1, emphasizing the need for vaccines that induce durable sterilizing immunity. Here we optimized regimens for chemoprophylaxis vaccination (CVac), for which aseptic, purified, cryopreserved, infectious Plasmodium falciparum sporozoites (PfSPZ) were inoculated under prophylactic cover with pyrimethamine (PYR) (Sanaria PfSPZ-CVac(PYR)) or chloroquine (CQ) (PfSPZ-CVac(CQ))-which kill liver-stage and blood-stage parasites, respectively-and we assessed vaccine efficacy against homologous (that is, the same strain as the vaccine) and heterologous (a different strain) controlled human malaria infection (CHMI) three months after immunization ( https://clinicaltrials.gov/ , NCT02511054 and NCT03083847). We report that a fourfold increase in the dose of PfSPZ-CVac(PYR) from 5.12 × 104 to 2 × 105 PfSPZs transformed a minimal vaccine efficacy (low dose, two out of nine (22.2%) participants protected against homologous CHMI), to a high-level vaccine efficacy with seven out of eight (87.5%) individuals protected against homologous and seven out of nine (77.8%) protected against heterologous CHMI. Increased protection was associated with Vδ2 γδ T cell and antibody responses. At the higher dose, PfSPZ-CVac(CQ) protected six out of six (100%) participants against heterologous CHMI three months after immunization. All homologous (four out of four) and heterologous (eight out of eight) infectivity control participants showed parasitaemia. PfSPZ-CVac(CQ) and PfSPZ-CVac(PYR) induced a durable, sterile vaccine efficacy against a heterologous South American strain of P. falciparum, which has a genome and predicted CD8 T cell immunome that differs more strongly from the African vaccine strain than other analysed African P. falciparum strains.


Antibodies, Neutralizing/immunology , Liver/immunology , Liver/parasitology , Malaria Vaccines/immunology , Plasmodium falciparum/drug effects , Plasmodium falciparum/immunology , Vaccines, Attenuated/immunology , Adult , Animals , Antibody Formation/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Life Cycle Stages/immunology , Malaria/blood , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Malaria Vaccines/chemistry , Male , Middle Aged , Plasmodium falciparum/growth & development , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Vaccination/adverse effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/chemistry
5.
Sci Transl Med ; 12(553)2020 07 22.
Article En | MEDLINE | ID: mdl-32718991

Vaccine development has the potential to be accelerated by coupling tools such as systems immunology analyses and controlled human infection models to define the protective efficacy of prospective immunogens without expensive and slow phase 2b/3 vaccine studies. Among human challenge models, controlled human malaria infection trials have long been used to evaluate candidate vaccines, and RTS,S/AS01 is the most advanced malaria vaccine candidate, reproducibly demonstrating 40 to 80% protection in human challenge studies in malaria-naïve individuals. Although antibodies are critical for protection after RTS,S/AS01 vaccination, antibody concentrations are inconsistently associated with protection across studies, and the precise mechanism(s) by which vaccine-induced antibodies provide protection remains enigmatic. Using a comprehensive systems serological profiling platform, the humoral correlates of protection against malaria were identified and validated across multiple challenge studies. Rather than antibody concentration, qualitative functional humoral features robustly predicted protection from infection across vaccine regimens. Despite the functional diversity of vaccine-induced immune responses across additional RTS,S/AS01 vaccine studies, the same antibody features, antibody-mediated phagocytosis and engagement of Fc gamma receptor 3A (FCGR3A), were able to predict protection across two additional human challenge studies. Functional validation using monoclonal antibodies confirmed the protective role of Fc-mediated antibody functions in restricting parasite infection both in vitro and in vivo, suggesting that these correlates may mechanistically contribute to parasite restriction and can be used to guide the rational design of an improved vaccine against malaria.


Malaria Vaccines , Malaria, Falciparum , Malaria , Antibodies, Protozoan , Humans , Malaria/prevention & control , Malaria, Falciparum/prevention & control , Plasmodium falciparum , Prospective Studies , Receptors, IgG , Vaccination
6.
Nat Commun ; 10(1): 3226, 2019 07 19.
Article En | MEDLINE | ID: mdl-31324806

Primaquine (PQ) is an essential antimalarial drug but despite being developed over 70 years ago, its mode of action is unclear. Here, we demonstrate that hydroxylated-PQ metabolites (OH-PQm) are responsible for efficacy against liver and sexual transmission stages of Plasmodium falciparum. The antimalarial activity of PQ against liver stages depends on host CYP2D6 status, whilst OH-PQm display direct, CYP2D6-independent, activity. PQ requires hepatic metabolism to exert activity against gametocyte stages. OH-PQm exert modest antimalarial efficacy against parasite gametocytes; however, potency is enhanced ca.1000 fold in the presence of cytochrome P450 NADPH:oxidoreductase (CPR) from the liver and bone marrow. Enhancement of OH-PQm efficacy is due to the direct reduction of quinoneimine metabolites by CPR with the concomitant and excessive generation of H2O2, leading to parasite killing. This detailed understanding of the mechanism paves the way to rationally re-designed 8-aminoquinolines with improved pharmacological profiles.


Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Primaquine/metabolism , Primaquine/pharmacology , Aminoquinolines/pharmacology , Bone Marrow/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 Enzyme System , Dose-Response Relationship, Drug , Humans , Hydrogen Peroxide/metabolism , Liver/metabolism , Malaria, Falciparum/drug therapy , NADP , Pharmacokinetics
7.
Cell ; 177(2): 315-325.e14, 2019 04 04.
Article En | MEDLINE | ID: mdl-30929905

Transmission of malaria parasites occurs when a female Anopheles mosquito feeds on an infected host to acquire nutrients for egg development. How parasites are affected by oogenetic processes, principally orchestrated by the steroid hormone 20-hydroxyecdysone (20E), remains largely unknown. Here we show that Plasmodium falciparum development is intimately but not competitively linked to processes shaping Anopheles gambiae reproduction. We unveil a 20E-mediated positive correlation between egg and oocyst numbers; impairing oogenesis by multiple 20E manipulations decreases parasite intensities. These manipulations, however, accelerate Plasmodium growth rates, allowing sporozoites to become infectious sooner. Parasites exploit mosquito lipids for faster growth, but they do so without further affecting egg development. These results suggest that P. falciparum has adopted a non-competitive evolutionary strategy of resource exploitation to optimize transmission while minimizing fitness costs to its mosquito vector. Our findings have profound implications for currently proposed control strategies aimed at suppressing mosquito populations.


Ecdysterone/metabolism , Host-Parasite Interactions/physiology , Malaria, Falciparum/parasitology , Animals , Anopheles/parasitology , Culicidae , Ecdysterone/physiology , Female , HEK293 Cells , Humans , Insect Vectors , Malaria/parasitology , Mice , Mosquito Vectors , NIH 3T3 Cells , Oogenesis/physiology , Plasmodium/metabolism , Plasmodium falciparum , Sporozoites , Steroids/metabolism
9.
Methods Mol Biol ; 1787: 41-52, 2018.
Article En | MEDLINE | ID: mdl-29736708

Malaria remains one of the deadliest infectious diseases globally. Available therapeutic agents are already limited in their efficacy, and drug resistance threatens to diminish further our ability to prevent and treat the disease. Despite a renewed effort to identify compounds with antimalarial activity, the drug discovery and development pipeline lacks target diversity and availability of compounds that target liver- and gametocyte-stage parasites. Phenotypic screens are a powerful and valuable tool for identifying new chemical compounds with antimalarial activity. This chapter highlights recent phenotypic screening methodologies for all three parasitic life stages.


Antimalarials/pharmacology , Drug Discovery , Life Cycle Stages/drug effects , Plasmodium/drug effects , Plasmodium/physiology , Small Molecule Libraries , Erythrocytes/drug effects , Erythrocytes/parasitology , Humans , Liver/drug effects , Liver/parasitology , Phenotype
10.
Nat Med ; 24(4): 408-416, 2018 05.
Article En | MEDLINE | ID: mdl-29554083

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.


Malaria Vaccines/immunology , Malaria/immunology , Parasites/immunology , Protozoan Proteins/chemistry , Animals , Antibodies, Monoclonal , Antibodies, Protozoan/immunology , Humans , Mice , Plasmodium falciparum/immunology
11.
Cell Host Microbe ; 23(3): 395-406.e4, 2018 Mar 14.
Article En | MEDLINE | ID: mdl-29478773

The unique relapsing nature of Plasmodium vivax infection is a major barrier to malaria eradication. Upon infection, dormant liver-stage forms, hypnozoites, linger for weeks to months and then relapse to cause recurrent blood-stage infection. Very little is known about hypnozoite biology; definitive biomarkers are lacking and in vitro platforms that support phenotypic studies are needed. Here, we recapitulate the entire liver stage of P. vivax in vitro, using a multiwell format that incorporates micropatterned primary human hepatocyte co-cultures (MPCCs). MPCCs feature key aspects of P. vivax biology, including establishment of persistent small forms and growing schizonts, merosome release, and subsequent infection of reticulocytes. We find that the small forms exhibit previously described hallmarks of hypnozoites, and we pilot MPCCs as a tool for testing candidate anti-hypnozoite drugs. Finally, we employ a hybrid capture strategy and RNA sequencing to describe the hypnozoite transcriptome and gain insight into its biology.


Antimalarials/pharmacology , Cell Culture Techniques/methods , Parasitic Sensitivity Tests/methods , Plasmodium vivax/drug effects , Plasmodium vivax/growth & development , Plasmodium vivax/metabolism , Transcriptome , Animals , Biomarkers , Cell Line/parasitology , Coculture Techniques , Fibroblasts , Hepatocytes/parasitology , Humans , In Vitro Techniques , Kinetics , Liver/parasitology , Malaria, Vivax/drug therapy , Mice , Sequence Analysis, RNA , Sporozoites/drug effects , Sporozoites/growth & development , Sporozoites/metabolism
12.
Sci Rep ; 7: 45424, 2017 03 31.
Article En | MEDLINE | ID: mdl-28361899

The malaria liver stage is an attractive target for antimalarial development, and preclinical malaria models are essential for testing such candidates. Given ethical concerns and costs associated with non-human primate models, humanized mouse models containing chimeric human livers offer a valuable alternative as small animal models of liver stage human malaria. The best available human liver chimeric mice rely on cellular transplantation into mice with genetically engineered liver injury, but these systems involve a long and variable humanization process, are expensive, and require the use of breeding-challenged mouse strains which are not widely accessible. We previously incorporated primary human hepatocytes into engineered polyethylene glycol (PEG)-based nanoporous human ectopic artificial livers (HEALs), implanted them in mice without liver injury, and rapidly generated human liver chimeric mice in a reproducible and scalable fashion. By re-designing the PEG scaffold to be macroporous, we demonstrate the facile fabrication of implantable porous HEALs that support liver stage human malaria (P. falciparum) infection in vitro, and also after implantation in mice with normal liver function, 60% of the time. This proof-of-concept study demonstrates the feasibility of applying a tissue engineering strategy towards the development of scalable preclinical models of liver stage malaria infection for future applications.


Disease Models, Animal , Liver/parasitology , Malaria/pathology , Malaria/parasitology , Animals , Cell Transplantation/methods , Humans , Mice
13.
Nature ; 538(7625): 344-349, 2016 Oct 20.
Article En | MEDLINE | ID: mdl-27602946

Antimalarial drugs have thus far been chiefly derived from two sources-natural products and synthetic drug-like compounds. Here we investigate whether antimalarial agents with novel mechanisms of action could be discovered using a diverse collection of synthetic compounds that have three-dimensional features reminiscent of natural products and are underrepresented in typical screening collections. We report the identification of such compounds with both previously reported and undescribed mechanisms of action, including a series of bicyclic azetidines that inhibit a new antimalarial target, phenylalanyl-tRNA synthetase. These molecules are curative in mice at a single, low dose and show activity against all parasite life stages in multiple in vivo efficacy models. Our findings identify bicyclic azetidines with the potential to both cure and prevent transmission of the disease as well as protect at-risk populations with a single oral dose, highlighting the strength of diversity-oriented synthesis in revealing promising therapeutic targets.


Antimalarials/chemical synthesis , Antimalarials/pharmacology , Azetidines/therapeutic use , Drug Discovery , Life Cycle Stages/drug effects , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Animals , Antimalarials/administration & dosage , Antimalarials/therapeutic use , Azabicyclo Compounds/administration & dosage , Azabicyclo Compounds/chemical synthesis , Azabicyclo Compounds/pharmacology , Azabicyclo Compounds/therapeutic use , Azetidines/administration & dosage , Azetidines/adverse effects , Azetidines/pharmacology , Cytosol/enzymology , Disease Models, Animal , Female , Liver/drug effects , Liver/parasitology , Macaca mulatta/parasitology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Male , Mice , Phenylalanine-tRNA Ligase/antagonists & inhibitors , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Plasmodium falciparum/cytology , Plasmodium falciparum/enzymology , Safety
14.
Int J Parasitol ; 46(11): 679-83, 2016 10.
Article En | MEDLINE | ID: mdl-27456552

Plasmodium vivax is the most geographically widespread malaria parasite. Unique features of transmission biology complicate P. vivax control. Interventions targeting transmission are required for malaria eradication. In the absence of an in vitro culture, transmission studies rely on live isolates from non-human primates or endemic regions. Here, we demonstrate P. vivax gametocytes from both India and Brazil are stable during cryopreservation. Importantly, cryopreserved gametocytes from Brazil were capable of infecting three anopheline mosquito species in feedings done in the United States. These findings create new opportunities for transmission studies in diverse locales.


Anopheles/parasitology , Cryopreservation , Insect Vectors/parasitology , Malaria, Vivax/parasitology , Plasmodium vivax/physiology , Animals , Brazil , Humans , India , Malaria, Vivax/transmission
15.
Nat Protoc ; 10(12): 2027-53, 2015 Dec.
Article En | MEDLINE | ID: mdl-26584444

The development of therapies and vaccines for human hepatropic pathogens requires robust model systems that enable the study of host-pathogen interactions. However, in vitro liver models of infection typically use either hepatoma cell lines that exhibit aberrant physiology or primary human hepatocytes in culture conditions in which they rapidly lose their hepatic phenotype. To achieve stable and robust in vitro primary human hepatocyte models, we developed micropatterned cocultures (MPCCs), which consist of primary human hepatocytes organized into 2D islands that are surrounded by supportive fibroblast cells. By using this system, which can be established over a period of days, and maintained over multiple weeks, we demonstrate how to recapitulate in vitro hepatic life cycles for the hepatitis B and C viruses and the Plasmodium pathogens P. falciparum and P. vivax. The MPCC platform can be used to uncover aspects of host-pathogen interactions, and it has the potential to be used for drug and vaccine development.


Coculture Techniques/methods , Hepacivirus/physiology , Hepatitis B virus/physiology , Hepatocytes/parasitology , Hepatocytes/virology , Host-Pathogen Interactions , Plasmodium/physiology , Animals , Cell Line , Cells, Cultured , Fibroblasts/cytology , Hepatitis B/metabolism , Hepatitis C/metabolism , Hepatocytes/cytology , Humans , Malaria/metabolism , Mice , Plasmodium falciparum/physiology , Plasmodium vivax/physiology , Tissue Array Analysis/methods
16.
Sci Transl Med ; 7(296): 296ra111, 2015 Jul 15.
Article En | MEDLINE | ID: mdl-26180101

Malaria is one of the most significant causes of childhood mortality, but disease control efforts are threatened by resistance of the Plasmodium parasite to current therapies. Continued progress in combating malaria requires development of new, easy to administer drug combinations with broad-ranging activity against all manifestations of the disease. DSM265, a triazolopyrimidine-based inhibitor of the pyrimidine biosynthetic enzyme dihydroorotate dehydrogenase (DHODH), is the first DHODH inhibitor to reach clinical development for treatment of malaria. We describe studies profiling the biological activity, pharmacological and pharmacokinetic properties, and safety of DSM265, which supported its advancement to human trials. DSM265 is highly selective toward DHODH of the malaria parasite Plasmodium, efficacious against both blood and liver stages of P. falciparum, and active against drug-resistant parasite isolates. Favorable pharmacokinetic properties of DSM265 are predicted to provide therapeutic concentrations for more than 8 days after a single oral dose in the range of 200 to 400 mg. DSM265 was well tolerated in repeat-dose and cardiovascular safety studies in mice and dogs, was not mutagenic, and was inactive against panels of human enzymes/receptors. The excellent safety profile, blood- and liver-stage activity, and predicted long half-life in humans position DSM265 as a new potential drug combination partner for either single-dose treatment or once-weekly chemoprevention. DSM265 has advantages over current treatment options that are dosed daily or are inactive against the parasite liver stage.


Antimalarials/chemistry , Enzyme Inhibitors/chemistry , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Pyrimidines/chemistry , Triazoles/chemistry , Administration, Oral , Animals , Antimalarials/pharmacokinetics , Area Under Curve , Caco-2 Cells , Crystallography, X-Ray , Dihydroorotate Dehydrogenase , Dogs , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Haplorhini , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Plasmodium falciparum , Pyrimidines/pharmacokinetics , Rabbits , Substrate Specificity , Triazoles/pharmacokinetics
17.
Stem Cell Reports ; 4(3): 348-59, 2015 Mar 10.
Article En | MEDLINE | ID: mdl-25660406

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.


Cell Differentiation , Hepatocytes/cytology , Hepatocytes/parasitology , Induced Pluripotent Stem Cells/cytology , Plasmodium/physiology , Antimalarials/pharmacology , Cells, Cultured , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , In Vitro Techniques , Malaria/parasitology
18.
Eukaryot Cell ; 14(1): 96-103, 2015 Jan.
Article En | MEDLINE | ID: mdl-25416236

Prior to invading nonreplicative erythrocytes, Plasmodium parasites undergo their first obligate step in the mammalian host inside hepatocytes, where each sporozoite replicates to generate thousands of merozoites. While normally quiescent, hepatocytes retain proliferative capacity and can readily reenter the cell cycle in response to diverse stimuli. Many intracellular pathogens, including protozoan parasites, manipulate the cell cycle progression of their host cells for their own benefit, but it is not known whether the hepatocyte cell cycle plays a role during Plasmodium liver stage infection. Here, we show that Plasmodium parasites can be observed in mitotic hepatoma cells throughout liver stage development, where they initially reduce the likelihood of mitosis and ultimately lead to significant acquisition of a binucleate phenotype. However, hepatoma cells pharmacologically arrested in S phase still support robust and complete Plasmodium liver stage development, which thus does not require cell cycle progression in the infected cell in vitro. Furthermore, murine hepatocytes remain quiescent throughout in vivo infection with either Plasmodium berghei or Plasmodium yoelii, as do Plasmodium falciparum-infected primary human hepatocytes, demonstrating that the rapid and prodigious growth of liver stage parasites is accomplished independent of host hepatocyte cell cycle progression during natural infection.


Hepatocytes/physiology , Mitosis , Plasmodium/growth & development , Sporozoites/physiology , Animals , Cell Nucleus/physiology , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/parasitology , Host-Parasite Interactions , Humans , Mice , Mice, Inbred C57BL , Plasmodium/pathogenicity
19.
Cell Host Microbe ; 16(6): 778-86, 2014 Dec 10.
Article En | MEDLINE | ID: mdl-25498345

During invasion, Plasmodium, the causative agent of malaria, wraps itself in a parasitophorous vacuole membrane (PVM), which constitutes a critical interface between the parasite and its host cell. Within hepatocytes, each Plasmodium sporozoite generates thousands of new parasites, creating high demand for lipids to support this replication and enlarge the PVM. Here, a global analysis of the total lipid repertoire of Plasmodium-infected hepatocytes reveals an enrichment of neutral lipids and the major membrane phospholipid, phosphatidylcholine (PC). While infection is unaffected in mice deficient in key enzymes involved in neutral lipid synthesis and lipolysis, ablation of rate-limiting enzymes in hepatic PC biosynthetic pathways significantly decreases parasite numbers. Host PC is taken up by both P. berghei and P. falciparum and is necessary for correct localization of parasite proteins to the PVM, which is essential for parasite survival. Thus, Plasmodium relies on the abundance of these lipids within hepatocytes to support infection.


Liver/parasitology , Malaria/metabolism , Phosphatidylcholines/biosynthesis , Plasmodium berghei/metabolism , Plasmodium falciparum/metabolism , Animals , Cell Line , Cell Survival , Female , Host-Parasite Interactions , Humans , Lipid Metabolism , Liver/metabolism , Malaria/parasitology , Mice , Mice, Inbred C57BL , Plasmodium berghei/growth & development , Plasmodium falciparum/growth & development , Sporozoites/growth & development , Sporozoites/metabolism
20.
Dis Model Mech ; 7(2): 215-24, 2014 Feb.
Article En | MEDLINE | ID: mdl-24291761

Homeostasis of mammalian cell function strictly depends on balancing oxygen exposure to maintain energy metabolism without producing excessive reactive oxygen species. In vivo, cells in different tissues are exposed to a wide range of oxygen concentrations, and yet in vitro models almost exclusively expose cultured cells to higher, atmospheric oxygen levels. Existing models of liver-stage malaria that utilize primary human hepatocytes typically exhibit low in vitro infection efficiencies, possibly due to missing microenvironmental support signals. One cue that could influence the infection capacity of cultured human hepatocytes is the dissolved oxygen concentration. We developed a microscale human liver platform comprised of precisely patterned primary human hepatocytes and nonparenchymal cells to model liver-stage malaria, but the oxygen concentrations are typically higher in the in vitro liver platform than anywhere along the hepatic sinusoid. Indeed, we observed that liver-stage Plasmodium parasite development in vivo correlates with hepatic sinusoidal oxygen gradients. Therefore, we hypothesized that in vitro liver-stage malaria infection efficiencies might improve under hypoxia. Using the infection of micropatterned co-cultures with Plasmodium berghei, Plasmodium yoelii or Plasmodium falciparum as a model, we observed that ambient hypoxia resulted in increased survival of exo-erythrocytic forms (EEFs) in hepatocytes and improved parasite development in a subset of surviving EEFs, based on EEF size. Further, the effective cell surface oxygen tensions (pO2) experienced by the hepatocytes, as predicted by a mathematical model, were systematically perturbed by varying culture parameters such as hepatocyte density and height of the medium, uncovering an optimal cell surface pO2 to maximize the number of mature EEFs. Initial mechanistic experiments revealed that treatment of primary human hepatocytes with the hypoxia mimetic, cobalt(II) chloride, as well as a HIF-1α activator, dimethyloxalylglycine, also enhance P. berghei infection, suggesting that the effect of hypoxia on infection is mediated in part by host-dependent HIF-1α mechanisms.


Hepatocytes/parasitology , Hypoxia/complications , Life Cycle Stages , Liver/pathology , Liver/parasitology , Malaria/complications , Malaria/parasitology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Coculture Techniques , Erythrocytes/drug effects , Erythrocytes/parasitology , Erythrocytes/pathology , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Hypoxia/parasitology , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kinetics , Liver/drug effects , Malaria/pathology , Mice , Oxygen/pharmacology , Partial Pressure , Plasmodium/drug effects , Plasmodium/growth & development , Sporozoites/drug effects , Sporozoites/physiology , Survival Analysis
...