Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Host Microbe ; 23(3): 395-406.e4, 2018 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-29478773

RESUMO

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.


Assuntos
Antimaláricos/farmacologia , Técnicas de Cultura de Células/métodos , Testes de Sensibilidade Parasitária/métodos , Plasmodium vivax/efeitos dos fármacos , Plasmodium vivax/crescimento & desenvolvimento , Plasmodium vivax/metabolismo , Transcriptoma , Animais , Biomarcadores , Linhagem Celular/parasitologia , Técnicas de Cocultura , Fibroblastos , Hepatócitos/parasitologia , Humanos , Técnicas In Vitro , Cinética , Fígado/parasitologia , Malária Vivax/tratamento farmacológico , Camundongos , Análise de Sequência de RNA , Esporozoítos/efeitos dos fármacos , Esporozoítos/crescimento & desenvolvimento , Esporozoítos/metabolismo
2.
Sci Rep ; 7: 45424, 2017 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-28361899

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Fígado/parasitologia , Malária/patologia , Malária/parasitologia , Animais , Transplante de Células/métodos , Humanos , Camundongos
3.
Nat Protoc ; 10(12): 2027-53, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26584444

RESUMO

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.


Assuntos
Técnicas de Cocultura/métodos , Hepacivirus/fisiologia , Vírus da Hepatite B/fisiologia , Hepatócitos/parasitologia , Hepatócitos/virologia , Interações Hospedeiro-Patógeno , Plasmodium/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Fibroblastos/citologia , Hepatite B/metabolismo , Hepatite C/metabolismo , Hepatócitos/citologia , Humanos , Malária/metabolismo , Camundongos , Plasmodium falciparum/fisiologia , Plasmodium vivax/fisiologia , Análise Serial de Tecidos/métodos
4.
Stem Cell Reports ; 4(3): 348-59, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25660406

RESUMO

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.


Assuntos
Diferenciação Celular , Hepatócitos/citologia , Hepatócitos/parasitologia , Células-Tronco Pluripotentes Induzidas/citologia , Plasmodium/fisiologia , Antimaláricos/farmacologia , Células Cultivadas , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Técnicas In Vitro , Malária/parasitologia
5.
Dis Model Mech ; 7(2): 215-24, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24291761

RESUMO

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.


Assuntos
Hepatócitos/parasitologia , Hipóxia/complicações , Estágios do Ciclo de Vida , Fígado/patologia , Fígado/parasitologia , Malária/complicações , Malária/parasitologia , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Técnicas de Cocultura , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Eritrócitos/patologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Humanos , Hipóxia/parasitologia , Hipóxia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Cinética , Fígado/efeitos dos fármacos , Malária/patologia , Camundongos , Oxigênio/farmacologia , Pressão Parcial , Plasmodium/efeitos dos fármacos , Plasmodium/crescimento & desenvolvimento , Esporozoítos/efeitos dos fármacos , Esporozoítos/fisiologia , Análise de Sobrevida
6.
Cell Host Microbe ; 14(1): 104-15, 2013 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-23870318

RESUMO

The Plasmodium liver stage is an attractive target for the development of antimalarial drugs and vaccines, as it provides an opportunity to interrupt the life cycle of the parasite at a critical early stage. However, targeting the liver stage has been difficult. Undoubtedly, a major barrier has been the lack of robust, reliable, and reproducible in vitro liver-stage cultures. Here, we establish the liver stages for both Plasmodium falciparum and Plasmodium vivax in a microscale human liver platform composed of cryopreserved, micropatterned human primary hepatocytes surrounded by supportive stromal cells. Using this system, we have successfully recapitulated the full liver stage of P. falciparum, including the release of infected merozoites and infection of overlaid erythrocytes, as well as the establishment of small forms in late liver stages of P. vivax. Finally, we validate the potential of this platform as a tool for medium-throughput antimalarial drug screening and vaccine development.


Assuntos
Hepatócitos/parasitologia , Fígado/citologia , Malária/parasitologia , Parasitologia/métodos , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium vivax/crescimento & desenvolvimento , Animais , Antimaláricos/farmacologia , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Hepatócitos/citologia , Humanos , Estágios do Ciclo de Vida , Fígado/parasitologia , Malária/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Plasmodium vivax/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...