1
|
Sefik E, Xiao T, Chiorazzi M, Odell I, Zhang F, Agrawal K, Micevic G, Flavell RA. Engineering Mice to Study Human Immunity. Annu Rev Immunol 2025; 43:451-487. [PMID: 40020225 DOI: 10.1146/annurev-immunol-082523-124415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Humanized mice, which carry a human hematopoietic and immune system, have greatly advanced our understanding of human immune responses and immunological diseases. These mice are created via the transplantation of human hematopoietic stem and progenitor cells into immunocompromised murine hosts further engineered to support human hematopoiesis and immune cell growth. This article explores genetic modifications in mice that enhance xeno-tolerance, promote human hematopoiesis and immunity, and enable xenotransplantation of human tissues with resident immune cells. We also discuss genetic editing of the human immune system, provide examples of how humanized mice with humanized organs model diseases for mechanistic studies, and highlight the roles of these models in advancing knowledge of organ biology, immune responses to pathogens, and preclinical drugs tested for cancer treatment. The integration of multi-omics and state-of-the art approaches with humanized mouse models is crucial for bridging existing human data with causality and promises to significantly advance mechanistic studies.
Collapse
Affiliation(s)
- Esen Sefik
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
| | - Tianli Xiao
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael Chiorazzi
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ian Odell
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Fengrui Zhang
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kriti Agrawal
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Computational Biology and Bioinformatics Program, Yale University, New Haven, Connecticut, USA
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Goran Micevic
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; ,
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
2
|
Obaldía N. The human malaria- Aotus monkey model: a historical perspective in antimalarial chemotherapy research at the Gorgas Memorial Laboratory-Panama. Antimicrob Agents Chemother 2024; 68:e0033824. [PMID: 38837364 PMCID: PMC11232403 DOI: 10.1128/aac.00338-24] [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] [Indexed: 06/07/2024] Open
Abstract
The human malaria-Aotus monkey model has served the malaria research community since its inception in 1966 at the Gorgas Memorial Laboratory (GML) in Panama. Spanning over five decades, this model has been instrumental in evaluating the in vivo efficacy and pharmacokinetics of a wide array of candidate antimalarial drugs, whether used singly or in combination. The animal model could be infected with drug-resistant and susceptible Plasmodium falciparum and Plasmodium vivax strains that follow a characteristic and reproducible course of infection, remarkably like human untreated and treated infections. Over the years, the model has enabled the evaluation of several synthetic and semisynthetic endoperoxides, for instance, artelinic acid, artesunate, artemether, arteether, and artemisone. These compounds have been evaluated alone and in combination with long-acting partner drugs, commonly referred to as artemisinin-based combination therapies, which are recommended as first-line treatment against uncomplicated malaria. Further, the model has also supported the evaluation of the primaquine analog tafenoquine against blood stages of P. vivax, contributing to its progression to clinical trials and eventual approval. Besides, the P. falciparum/Aotus model at GML has also played a pivotal role in exploring the biology, immunology, and pathogenesis of malaria and in the characterization of drug-resistant P. falciparum and P. vivax strains. This minireview offers a historical overview of the most significant contributions made by the Panamanian owl monkey (Aotus lemurinus lemurinus) to malaria chemotherapy research.
Collapse
Affiliation(s)
- Nicanor Obaldía
- Center for the Evaluation of Antimalarial Drugs and Vaccines, Instituto Conmemorativo Gorgas de Estudios de la Salud, Panama, Republic of Panama
- Department of Immunology and Infectious Diseases, Harvard University T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Nicholas J, Kolli SK, Subramani PA, De SL, Ogbondah MM, Barnes SJ, Ntumngia FB, Adams JH. Comparative analyses of functional antibody-mediated inhibition with anti-circumsporozoite monoclonal antibodies against transgenic Plasmodium berghei. Malar J 2023; 22:335. [PMID: 37936181 PMCID: PMC10629016 DOI: 10.1186/s12936-023-04765-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Acquired functional inhibitory antibodies are one of several humoral immune mechanisms used to neutralize foreign pathogens. In vitro bioassays are useful tools for quantifying antibody-mediated inhibition and evaluating anti-parasite immune antibodies. However, a gap remains in understanding of how antibody-mediated inhibition in vitro translates to inhibition in vivo. In this study, two well-characterized transgenic Plasmodium berghei parasite lines, PbmCh-luc and Pb-PfCSP(r), and murine monoclonal antibodies (mAbs) specific to P. berghei and Plasmodium falciparum circumsporozoite protein (CSP), 3D11 and 2A10, respectively, were used to evaluate antibody-mediated inhibition of parasite development in both in vitro and in vivo functional assays. METHODS IC50 values of mAbs were determined using an established inhibition of liver-stage development assay (ILSDA). For the in vivo inhibition assay, mice were passively immunized by transfer of the mAbs and subsequently challenged with 5.0 × 103 sporozoites via tail vein injection. The infection burden in both assays was quantified by luminescence and qRT-PCR of P. berghei 18S rRNA normalized to host GAPDH. RESULTS The IC50 values quantified by relative luminescence of mAbs 3D11 and 2A10 were 0.396 µg/ml and 0.093 µg/ml, respectively, against transgenic lines in vitro. Using the highest (> 90%) inhibitory antibody concentrations in a passive transfer, an IC50 of 233.8 µg/ml and 181.5 µg/ml for mAbs 3D11 and 2A10, respectively, was observed in vivo. At 25 µg (250 µg/ml), the 2A10 antibody significantly inhibited liver burden in mice compared to control. Additionally, qRT-PCR of P. berghei 18S rRNA served as a secondary validation of liver burden quantification. CONCLUSIONS Results from both experimental models, ILSDA and in vivo challenge, demonstrated that increased concentrations of the homologous anti-CSP repeat mAbs increased parasite inhibition. However, differences in antibody IC50 values between parasite lines did not allow a direct correlation between the inhibition of sporozoite invasion in vitro by ILSDA and the inhibition of mouse liver stage burden. Further studies are needed to establish the conditions for confident predictions for the in vitro ILSDA to be a predictor of in vivo outcomes using this model system.
Collapse
Affiliation(s)
- Justin Nicholas
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Surendra Kumar Kolli
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Pradeep Annamalai Subramani
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Sai Lata De
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
- Department of Infectious Disease & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Madison M Ogbondah
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Samantha J Barnes
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Francis Babila Ntumngia
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - John H Adams
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
4
|
Samayoa-Reyes G, Flaherty SM, Wickham KS, Viera-Morilla S, Strauch PM, Roth A, Padrón L, Jackson CM, Meireles P, Calvo D, Roobsoong W, Kangwanrangsan N, Sattabongkot J, Reichard G, Lafuente-Monasterio MJ, Rochford R. Development of an ectopic huLiver model for Plasmodium liver stage infection. PLoS One 2023; 18:e0279144. [PMID: 36928885 PMCID: PMC10019673 DOI: 10.1371/journal.pone.0279144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Early Plasmodium falciparum and P. vivax infection requires parasite replication within host hepatocytes, referred to as liver stage (LS). However, limited understanding of infection dynamics in human LS exists due to species-specificity challenges. Reported here is a reproducible, easy-to-manipulate, and moderate-cost in vivo model to study human Plasmodium LS in mice; the ectopic huLiver model. Ectopic huLiver tumors were generated through subcutaneous injection of the HC-04 cell line and shown to be infectible by both freshly dissected sporozoites and through the bite of infected mosquitoes. Evidence for complete LS development was supported by the transition to blood-stage infection in mice engrafted with human erythrocytes. Additionally, this model was successfully evaluated for its utility in testing antimalarial therapeutics, as supported by primaquine acting as a causal prophylactic against P. falciparum. Presented here is a new platform for the study of human Plasmodium infection with the potential to aid in drug discovery.
Collapse
Affiliation(s)
- Gabriela Samayoa-Reyes
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Siobhan M. Flaherty
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Kristina S. Wickham
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Sara Viera-Morilla
- Diseases of the Developing World, Infectious Diseases-Centre for Excellence in Drug Discovery (ID CEDD), GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Pamela M. Strauch
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Laura Padrón
- Diseases of the Developing World, Infectious Diseases-Centre for Excellence in Drug Discovery (ID CEDD), GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Conner M. Jackson
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Patricia Meireles
- Diseases of the Developing World, Infectious Diseases-Centre for Excellence in Drug Discovery (ID CEDD), GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - David Calvo
- Diseases of the Developing World, Infectious Diseases-Centre for Excellence in Drug Discovery (ID CEDD), GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Wanlapa Roobsoong
- Faculty of Tropical Medicine, Mahidol Vivax Research Unit, Mahidol University, Bangkok, Thailand
| | - Niwat Kangwanrangsan
- Faculty of Science, Pathobiology Department, Mahidol University, Bangkok, Thailand
| | - Jetsumon Sattabongkot
- Faculty of Tropical Medicine, Mahidol Vivax Research Unit, Mahidol University, Bangkok, Thailand
| | - Gregory Reichard
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Maria José Lafuente-Monasterio
- Diseases of the Developing World, Infectious Diseases-Centre for Excellence in Drug Discovery (ID CEDD), GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Rosemary Rochford
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
5
|
Valenciano AL, Gomez-Lorenzo MG, Vega-Rodríguez J, Adams JH, Roth A. In vitro models for human malaria: targeting the liver stage. Trends Parasitol 2022; 38:758-774. [PMID: 35780012 PMCID: PMC9378454 DOI: 10.1016/j.pt.2022.05.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
The Plasmodium liver stage represents a vulnerable therapeutic target to prevent disease progression as the parasite resides in the liver before clinical representation caused by intraerythrocytic development. However, most antimalarial drugs target the blood stage of the parasite's life cycle, and the few drugs that target the liver stage are lethal to patients with a glucose-6-phosphate dehydrogenase deficiency. Furthermore, implementation of in vitro liver models to study and develop novel therapeutics against the liver stage of human Plasmodium species remains challenging. In this review, we focus on the progression of in vitro liver models developed for human Plasmodium spp. parasites, provide a brief review on important assay requirements, and lastly present recommendations to improve models to enhance the discovery process of novel preclinical therapeutics.
Collapse
Affiliation(s)
- Ana Lisa Valenciano
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA; Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Maria G Gomez-Lorenzo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
6
|
Simwela NV, Waters AP. Current status of experimental models for the study of malaria. Parasitology 2022; 149:1-22. [PMID: 35357277 PMCID: PMC9378029 DOI: 10.1017/s0031182021002134] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Infection by malaria parasites (Plasmodium spp.) remains one of the leading causes of morbidity and mortality, especially in tropical regions of the world. Despite the availability of malaria control tools such as integrated vector management and effective therapeutics, these measures have been continuously undermined by the emergence of vector resistance to insecticides or parasite resistance to frontline antimalarial drugs. Whilst the recent pilot implementation of the RTS,S malaria vaccine is indeed a remarkable feat, highly effective vaccines against malaria remain elusive. The barriers to effective vaccines result from the complexity of both the malaria parasite lifecycle and the parasite as an organism itself with consequent major gaps in our understanding of their biology. Historically and due to the practical and ethical difficulties of working with human malaria infections, research into malaria parasite biology has been extensively facilitated by animal models. Animals have been used to study disease pathogenesis, host immune responses and their (dys)regulation and further disease processes such as transmission. Moreover, animal models remain at the forefront of pre-clinical evaluations of antimalarial drugs (drug efficacy, mode of action, mode of resistance) and vaccines. In this review, we discuss commonly used animal models of malaria, the parasite species used and their advantages and limitations which hinder their extrapolation to actual human disease. We also place into this context the most recent developments such as organoid technologies and humanized mice.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Andrew P. Waters
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| |
Collapse
|
7
|
Luo Y, Lu H, Peng D, Ruan X, Chen YE, Guo Y. Liver-humanized mice: A translational strategy to study metabolic disorders. J Cell Physiol 2022; 237:489-506. [PMID: 34661916 PMCID: PMC9126562 DOI: 10.1002/jcp.30610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/07/2021] [Accepted: 09/11/2021] [Indexed: 01/03/2023]
Abstract
The liver is the metabolic core of the whole body. Tools commonly used to study the human liver metabolism include hepatocyte cell lines, primary human hepatocytes, and pluripotent stem cells-derived hepatocytes in vitro, and liver genetically humanized mouse model in vivo. However, none of these systems can mimic the human liver in physiological and pathological states satisfactorily. Liver-humanized mice, which are established by reconstituting mouse liver with human hepatocytes, have emerged as an attractive animal model to study drug metabolism and evaluate the therapeutic effect in "human liver" in vivo because the humanized livers greatly replicate enzymatic features of human hepatocytes. The application of liver-humanized mice in studying metabolic disorders is relatively less common due to the largely uncertain replication of metabolic profiles compared to humans. Here, we summarize the metabolic characteristics and current application of liver-humanized mouse models in metabolic disorders that have been reported in the literature, trying to evaluate the pros and cons of using liver-humanized mice as novel mouse models to study metabolic disorders.
Collapse
Affiliation(s)
- Yonghong Luo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Haocheng Lu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiangbo Ruan
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins School of Medicine, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA
| | - Y. Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Center for Advanced Models and Translational Sciences and Therapeutics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yanhong Guo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
8
|
Tyagi RK. Plasmodium falciparum-infected humanized mice: a viable preclinical tool. Immunotherapy 2021; 13:1345-1353. [PMID: 34424053 DOI: 10.2217/imt-2021-0102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
Extensive research conducted on mouse-human chimeras has advanced our understanding on infectious diseases including the human-malaria parasite, Plasmodium falciparum. In vitro culture of asexual-blood stage infection of P. falciparum does not answer all questions related to parasitology, pharmacology and immunology, and complex life cycle, complicated genome, evolution of drug resistance and poor diagnosis makes it difficult to understand the patho-biology of parasite. Unavailability of effective-vaccine and issues of drug resistance advocates the use of human cell/tissues reconstituted immunodeficient-mice to P. falciparum. A number of immunodeficient-strains (TK/NOG, FRG/NOD, NOD/SCID/IL-2 receptor γ chain null, NOD severe combined immunodeficiency gamma [NSG] mouse and NOD.Rag1-/- IL2Rγ-/- [NRG; DRAG]) are used for humanization purposes. Additionally, human-hematopoietic stem cells (CD34 reconstituted-NSG [human immune system]) mice support the engraftment and repopulation of immune effecters to study systemic inflammatory diseases.
Collapse
Affiliation(s)
- Rajeev K Tyagi
- Division of Cell Biology & Immunology, Biomedical Parasitology & Nano-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh, 160036, India
| |
Collapse
|
9
|
Goodier MR, Wolf AS, Riley EM. Differentiation and adaptation of natural killer cells for anti-malarial immunity. Immunol Rev 2019; 293:25-37. [PMID: 31762040 DOI: 10.1111/imr.12798] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/07/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
Natural killer cells employ a diverse arsenal of effector mechanisms to target intracellular pathogens. Differentiation of natural killer (NK) cell activation pathways occurs along a continuum from reliance on innate pro-inflammatory cytokines and stress-induced host ligands through to interaction with signals derived from acquired immune responses. Importantly, the degree of functional differentiation of the NK cell lineage influences the magnitude and specificity of interactions with host cells infected with viruses, bacteria, fungi, and parasites. Individual humans possess a vast diversity of distinct NK cell clones, each with the capacity to vary along this functional differentiation pathway, which - when combined - results in unique individual responses to different infections. Here we summarize these NK cell differentiation events, review evidence for direct interaction of malaria-infected host cells with NK cells and assess how innate inflammatory signals induced by malaria parasite-associated molecular patterns influence the indirect activation and function of NK cells. Finally, we discuss evidence that anti-malarial immunity develops in parallel with advancing NK differentiation, coincident with a loss of reliance on inflammatory signals, and a refined capacity of NK cells to target malaria parasites more precisely, particularly through antibody-dependent mechanisms.
Collapse
Affiliation(s)
- Martin R Goodier
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Asia-Sophia Wolf
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.,Department of Infection and Immunity, University College London, London, UK
| | - Eleanor M Riley
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.,The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| |
Collapse
|
10
|
Sayed IM, Elkhawaga AA, El-Mokhtar MA. In vivo models for studying Hepatitis E virus infection; Updates and applications. Virus Res 2019; 274:197765. [PMID: 31563457 DOI: 10.1016/j.virusres.2019.197765] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 02/08/2023]
Abstract
Hepatitis E virus (HEV) is the most common cause of acute viral hepatitis globally. HEV belongs to the Hepeviridae family and at least five genotypes (gt) infect humans. Several animal species are reservoirs for different HEV strains, and they are the source of infection for humans. Some HEV strains are species specific, but other strains could cross species and infect many hosts. The study of HEV infection and pathogenesis was hampered due to the lack of an in vitro and in vivo robust model system. The cell culture system has been established for certain HEV strains, especially gt3 and 4, but gt1 strains replicate poorly in vitro. To date, animal models are the best tool for studying HEV infection. Non-human primates (NHPs) and pigs are the main animal models used for studying HEV infection, but ethical and financial concerns restrict the use of NHPs in research. Therefore, new small animal models have been developed which help more progress in HEV research. In this review, we give updates on the animal models used for studying HEV infection, focusing on the applicability of each model in studying different HEV infections, cross-species infection, virus-host interaction, evaluation of anti-HEV therapies and testing potential HEV vaccines.
Collapse
Affiliation(s)
- Ibrahim M Sayed
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, California, USA; Medical Microbiology and Immunology Department, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Amal A Elkhawaga
- Medical Microbiology and Immunology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Mohamed A El-Mokhtar
- Medical Microbiology and Immunology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
11
|
Goh YS, McGuire D, Rénia L. Vaccination With Sporozoites: Models and Correlates of Protection. Front Immunol 2019; 10:1227. [PMID: 31231377 PMCID: PMC6560154 DOI: 10.3389/fimmu.2019.01227] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Despite continuous efforts, the century-old goal of eradicating malaria still remains. Multiple control interventions need to be in place simultaneously to achieve this goal. In addition to effective control measures, drug therapies and insecticides, vaccines are critical to reduce mortality and morbidity. Hence, there are numerous studies investigating various malaria vaccine candidates. Most of the malaria vaccine candidates are subunit vaccines. However, they have shown limited efficacy in Phase II and III studies. To date, only whole parasite formulations have been shown to induce sterile immunity in human. In this article, we review and discuss the recent developments in vaccination with sporozoites and the mechanisms of protection involved.
Collapse
Affiliation(s)
- Yun Shan Goh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore
| | - Daniel McGuire
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
12
|
Tyagi RK, Tandel N, Deshpande R, Engelman RW, Patel SD, Tyagi P. Humanized Mice Are Instrumental to the Study of Plasmodium falciparum Infection. Front Immunol 2018; 9:2550. [PMID: 30631319 PMCID: PMC6315153 DOI: 10.3389/fimmu.2018.02550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/17/2018] [Indexed: 02/05/2023] Open
Abstract
Research using humanized mice has advanced our knowledge and understanding of human haematopoiesis, non-adaptive and adaptive immunity, autoimmunity, infectious disease, cancer biology, and regenerative medicine. Challenges posed by the human-malaria parasite Plasmodium falciparum include its complex life cycle, the evolution of drug resistance against anti-malarials, poor diagnosis, and a lack of effective vaccines. Advancements in genetically engineered and immunodeficient mouse strains, have allowed for studies of the asexual blood stage, exoerythrocytic stage and the transition from liver-to-blood stage infection, in a single vertebrate host. This review discusses the process of "humanization" of various immunodeficient/transgenic strains and their contribution to translational biomedical research. Our work reviews the strategies employed to overcome the remaining-limitations of the developed human-mouse chimera(s).
Collapse
Affiliation(s)
- Rajeev K. Tyagi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Biomedical parasitology Unit, Institute Pasteur, Paris, France
- Department of Global Health, College of Public Health, University of South Florida, Tampa, FL, United States
| | - Nikunj Tandel
- Institute of Science, Nirma University, Ahmedabad, India
| | | | - Robert W. Engelman
- Department of Pediatrics, Pathology and Cell Biology, University of South Florida, Tampa, FL, United States
| | | | - Priyanka Tyagi
- Department of Basic and Applied Sciences, School of Engineering, GD Goenka University, Gurgaon, India
| |
Collapse
|
13
|
Gualdrón-López M, Flannery EL, Kangwanrangsan N, Chuenchob V, Fernandez-Orth D, Segui-Barber J, Royo F, Falcón-Pérez JM, Fernandez-Becerra C, Lacerda MVG, Kappe SHI, Sattabongkot J, Gonzalez JR, Mikolajczak SA, Del Portillo HA. Characterization of Plasmodium vivax Proteins in Plasma-Derived Exosomes From Malaria-Infected Liver-Chimeric Humanized Mice. Front Microbiol 2018; 9:1271. [PMID: 29988527 PMCID: PMC6026661 DOI: 10.3389/fmicb.2018.01271] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 05/24/2018] [Indexed: 12/14/2022] Open
Abstract
Exosomes are extracellular vesicles of endocytic origin containing molecular signatures implying the cell of origin; thus, they offer a unique opportunity to discover biomarkers of disease. Plasmodium vivax, responsible for more than half of all malaria cases outside Africa, is a major obstacle in the goal of malaria elimination due to the presence of dormant liver stages (hypnozoites), which after the initial infection may reactivate to cause disease. Hypnozoite infection is asymptomatic and there are currently no diagnostic tools to detect their presence. The human liver-chimeric (FRG huHep) mouse is a robust P. vivax infection model for exo-erythrocytic development of liver stages, including hypnozoites. We studied the proteome of plasma-derived exosomes isolated from P. vivax infected FRG huHep mice with the objective of identifying liver-stage expressed parasite proteins indicative of infection. Proteomic analysis of these exosomes showed the presence of 290 and 234 proteins from mouse and human origin, respectively, including canonical exosomal markers. Human proteins include proteins previously detected in liver-derived exosomes, highlighting the potential of this chimeric mouse model to study plasma exosomes derived unequivocally from human hepatocytes. Noticeably, we identified 17 parasite proteins including enzymes, surface proteins, components of the endocytic pathway and translation machinery, as well as uncharacterized proteins. Western blot analysis validated the presence of human arginase-I and an uncharacterized P. vivax protein in plasma-derived exosomes. This study represents a proof-of-principle that plasma-derived exosomes from P. vivax infected FRG-huHep mice contain human hepatocyte and P. vivax proteins with the potential to unveil biological features of liver infection and identify biomarkers of hypnozoite infection.
Collapse
Affiliation(s)
- Melisa Gualdrón-López
- Instituto Salud Global, Hospital Clinic-Universitat de Barcelona, Barcelona, Spain.,Institute for Health Sciences Trias I Pujol, Barcelona, Spain
| | - Erika L Flannery
- Center for Infectious Disease Research, Seattle, WA, United States
| | - Niwat Kangwanrangsan
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Vorada Chuenchob
- Center for Infectious Disease Research, Seattle, WA, United States
| | | | - Joan Segui-Barber
- Instituto Salud Global, Hospital Clinic-Universitat de Barcelona, Barcelona, Spain
| | - Felix Royo
- Exosomes Laboratory, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERHD), Derio, Spain
| | - Juan M Falcón-Pérez
- Exosomes Laboratory, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERHD), Derio, Spain.,Metabolomics platform, CIC bioGUNE, CIBERehd, Derio, Spain.,IKERBASKE Basque Foundation for Science, Bilbao, Spain
| | - Carmen Fernandez-Becerra
- Instituto Salud Global, Hospital Clinic-Universitat de Barcelona, Barcelona, Spain.,Institute for Health Sciences Trias I Pujol, Barcelona, Spain
| | - Marcus V G Lacerda
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, Brazil.,Instituto Leônidas & Maria Deane, Manaus, Brazil
| | - Stefan H I Kappe
- Center for Infectious Disease Research, Seattle, WA, United States
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Juan R Gonzalez
- Instituto Salud Global, Hospital Clinic-Universitat de Barcelona, Barcelona, Spain
| | | | - Hernando A Del Portillo
- Instituto Salud Global, Hospital Clinic-Universitat de Barcelona, Barcelona, Spain.,Institute for Health Sciences Trias I Pujol, Barcelona, Spain.,Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| |
Collapse
|
14
|
Minkah NK, Schafer C, Kappe SHI. Humanized Mouse Models for the Study of Human Malaria Parasite Biology, Pathogenesis, and Immunity. Front Immunol 2018; 9:807. [PMID: 29725334 PMCID: PMC5917005 DOI: 10.3389/fimmu.2018.00807] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/03/2018] [Indexed: 12/25/2022] Open
Abstract
Malaria parasite infection continues to inflict extensive morbidity and mortality in resource-poor countries. The insufficiently understood parasite biology, continuously evolving drug resistance and the lack of an effective vaccine necessitate intensive research on human malaria parasites that can inform the development of new intervention tools. Humanized mouse models have been greatly improved over the last decade and enable the direct study of human malaria parasites in vivo in the laboratory. Nevertheless, no small animal model developed so far is capable of maintaining the complete life cycle of Plasmodium parasites that infect humans. The ultimate goal is to develop humanized mouse systems in which a Plasmodium infection closely reproduces all stages of a parasite infection in humans, including pre-erythrocytic infection, blood stage infection and its associated pathology, transmission as well as the human immune response to infection. Here, we discuss current humanized mouse models and the future directions that should be taken to develop next-generation models for human malaria parasite research.
Collapse
Affiliation(s)
- Nana K Minkah
- Center for Infectious Disease Research, Seattle, WA, United States
| | - Carola Schafer
- Center for Infectious Disease Research, Seattle, WA, United States
| | - Stefan H I Kappe
- Center for Infectious Disease Research, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
15
|
Dragovic SM, Agunbiade TA, Freudzon M, Yang J, Hastings AK, Schleicher TR, Zhou X, Craft S, Chuang YM, Gonzalez F, Li Y, Hrebikova G, Tripathi A, Mlambo G, Almeras L, Ploss A, Dimopoulos G, Fikrig E. Immunization with AgTRIO, a Protein in Anopheles Saliva, Contributes to Protection against Plasmodium Infection in Mice. Cell Host Microbe 2018; 23:523-535.e5. [PMID: 29649443 PMCID: PMC5998332 DOI: 10.1016/j.chom.2018.03.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/30/2017] [Accepted: 03/09/2018] [Indexed: 01/02/2023]
Abstract
Plasmodium infection begins with the bite of an anopheline mosquito, when sporozoites along with saliva are injected into a vertebrate host. The role of the host responses to mosquito saliva components in malaria remains unclear. We observed that antisera against Anopheles gambiae salivary glands partially protected mice from mosquito-borne Plasmodium infection. Specifically, antibodies to A. gambiae TRIO (AgTRIO), a mosquito salivary gland antigen, contributed to the protection. Mice administered AgTRIO antiserum showed lower Plasmodium liver burden and decreased parasitemia when exposed to infected mosquitoes. Active immunization with AgTRIO was also partially protective against Plasmodium berghei infection. A combination of AgTRIO antiserum and antibodies against Plasmodium circumsporozoite protein, a vaccine candidate, further decreased P. berghei infection. In humanized mice, AgTRIO antiserum afforded some protection against mosquito-transmitted Plasmodium falciparum. AgTRIO antiserum reduced the movement of sporozoites in the murine dermis. AgTRIO may serve as an arthropod-based target against Plasmodium to combat malaria.
Collapse
Affiliation(s)
- Srdjan M Dragovic
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA.
| | - Tolulope A Agunbiade
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA
| | - Marianna Freudzon
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA; Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jing Yang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA
| | - Andrew K Hastings
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA
| | - Tyler R Schleicher
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA
| | - Xia Zhou
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA
| | - Sam Craft
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA
| | - Yu-Min Chuang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA
| | - Floricel Gonzalez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA
| | - Youquan Li
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA
| | - Gabriela Hrebikova
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Abhai Tripathi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Godfree Mlambo
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lionel Almeras
- Unité de Parasitologie et Entomologie, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France; Aix Marseille Université, Marseille, France
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - George Dimopoulos
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, The Anlyan Center for Medical Research and Education, 300 Cedar Street, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
16
|
McCall MBB, Wammes LJ, Langenberg MCC, van Gemert GJ, Walk J, Hermsen CC, Graumans W, Koelewijn R, Franetich JF, Chishimba S, Gerdsen M, Lorthiois A, van de Vegte M, Mazier D, Bijker EM, van Hellemond JJ, van Genderen PJJ, Sauerwein RW. Infectivity of Plasmodium falciparum sporozoites determines emerging parasitemia in infected volunteers. Sci Transl Med 2018. [PMID: 28637923 DOI: 10.1126/scitranslmed.aag2490] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Malaria sporozoites must first undergo intrahepatic development before a pathogenic blood-stage infection is established. The success of infection depends on host and parasite factors. In healthy human volunteers undergoing controlled human malaria infection (CHMI), we directly compared three clinical Plasmodium falciparum isolates for their ability to infect primary human hepatocytes in vitro and to drive the production of blood-stage parasites in vivo. Our data show a correlation between the efficiency of strain-specific sporozoite invasion of human hepatocytes and the dynamics of patent parasitemia in study subjects, highlighting intrinsic differences in infectivity among P. falciparum isolates from distinct geographical locales. The observed heterogeneity in infectivity among strains underscores the value of assessing the protective efficacy of candidate malaria vaccines against heterologous strains in the CHMI model.
Collapse
Affiliation(s)
- Matthew B B McCall
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, Netherlands
| | - Linda J Wammes
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, Netherlands
| | | | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Jona Walk
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Cornelus C Hermsen
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Wouter Graumans
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Rob Koelewijn
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, Netherlands.,Institute for Tropical Diseases, Harbour Hospital, Rotterdam, Netherlands
| | - Jean-François Franetich
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France
| | - Sandra Chishimba
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, Netherlands
| | - Max Gerdsen
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Audrey Lorthiois
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France
| | - Marga van de Vegte
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Dominique Mazier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France.,AP-HP, Groupe hospitalier La Pitié-Salpêtrière, Service de Parasitologie Mycologie, F-75013 Paris, France
| | - Else M Bijker
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Jaap J van Hellemond
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, Netherlands
| | | | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands.
| |
Collapse
|
17
|
Abstract
Mice with humanized chimeric liver are promising in vivo tools to evaluate the efficacy of novel compounds or vaccine induced antibodies directed against pathogens that infect the human liver. In addition they can be used to study the human-type metabolism of medicinal compounds and hepatotoxicity.
Collapse
|
18
|
Moreno-Sabater A, Pérignon JL, Mazier D, Lavazec C, Soulard V. Humanized mouse models infected with human Plasmodium species for antimalarial drug discovery. Expert Opin Drug Discov 2017; 13:131-140. [DOI: 10.1080/17460441.2018.1410136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Alicia Moreno-Sabater
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
- Assistance Publique - Hopitaux de Paris - Hôpitaux Universitaires Paris-Est - Site Saint-Antoine, Paris, Île-de-France France
| | | | - Dominique Mazier
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
| | - Catherine Lavazec
- Institut Cochin – INSERM U1016, Paris, Île-de-France France
- CNRS - UMR8104, Paris, France
- Universite Paris Descartes, Paris, Île-de-France France
| | - Valerie Soulard
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
| |
Collapse
|
19
|
Gural N, Mancio-Silva L, He J, Bhatia SN. Engineered Livers for Infectious Diseases. Cell Mol Gastroenterol Hepatol 2017; 5:131-144. [PMID: 29322086 PMCID: PMC5756057 DOI: 10.1016/j.jcmgh.2017.11.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/02/2017] [Indexed: 01/18/2023]
Abstract
Engineered liver systems come in a variety of platform models, from 2-dimensional cocultures of primary human hepatocytes and stem cell-derived progeny, to 3-dimensional organoids and humanized mice. Because of the species-specificity of many human hepatropic pathogens, these engineered systems have been essential tools for biologic discovery and therapeutic agent development in the context of liver-dependent infectious diseases. Although improvement of existing models is always beneficial, and the addition of a robust immune component is a particular need, at present, considerable progress has been made using this combination of research platforms. We highlight advances in the study of hepatitis B and C viruses and malaria-causing Plasmodium falciparum and Plasmodium vivax parasites, and underscore the importance of pairing the most appropriate model system and readout modality with the particular experimental question at hand, without always requiring a platform that recapitulates human physiology in its entirety.
Collapse
Key Words
- 2D, 2-dimensional
- 3D
- 3D, 3-dimensional
- EBOV, Ebola virus
- Falciparum
- HBC, hepatitis C virus
- HBV
- HBV, hepatitis B virus
- HCV
- HLC, hepatocyte-like cells
- Hepatotropic
- LASV, Lassa virus
- Liver
- Liver Models
- MPCC, micropatterned coculture system
- Malaria
- PCR, polymerase chain reaction
- Pathogen
- SACC, self-assembling coculture
- Vivax
- iHLC, induced pluripotent stem cell–derived hepatocyte-like cells
- in vitro
- in vivo
Collapse
Affiliation(s)
- Nil Gural
- Harvard-MIT Department of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Boston, Massachusetts,Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Liliana Mancio-Silva
- Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Jiang He
- Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Sangeeta N. Bhatia
- Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts,Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts,Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts,Broad Institute, Cambridge, Massachusetts,Howard Hughes Medical Institute, Chevy Chase, Maryland,Correspondence Address correspondence to: Sangeeta N. Bhatia, MD, PhD, Koch Institute for Integrative Cancer, Research at MIT, Building 76, Room 473, 500 Main Street, Cambridge, Massachusetts 02142.
| |
Collapse
|
20
|
Armistead JS, Adams JH. Advancing Research Models and Technologies to Overcome Biological Barriers to Plasmodium vivax Control. Trends Parasitol 2017; 34:114-126. [PMID: 29153587 DOI: 10.1016/j.pt.2017.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 10/25/2017] [Accepted: 10/25/2017] [Indexed: 02/06/2023]
Abstract
Malaria prevalence has declined in the past 10 years, especially outside of sub-Saharan Africa. However, the proportion of cases due to Plasmodium vivax is increasing, accounting for up to 90-100% of the malaria burden in endemic regions. Nonetheless, investments in malaria research and control still prioritize Plasmodium falciparum while largely neglecting P. vivax. Specific biological features of P. vivax, particularly invasion of reticulocytes, occurrence of dormant liver forms of the parasite, and the potential for transmission of sexual-stage parasites prior to onset of clinical illness, promote its persistence and hinder development of research tools and interventions. This review discusses recent advances in P. vivax research, current knowledge of its unique biology, and proposes priorities for P. vivax research and control efforts.
Collapse
Affiliation(s)
- Jennifer S Armistead
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
21
|
Parker D. Humanized Mouse Models of Staphylococcus aureus Infection. Front Immunol 2017; 8:512. [PMID: 28523002 PMCID: PMC5415562 DOI: 10.3389/fimmu.2017.00512] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/18/2017] [Indexed: 12/18/2022] Open
Abstract
Staphylococcus aureus is a successful human pathogen that has adapted itself in response to selection pressure by the human immune system. A commensal of the human skin and nose, it is a leading cause of several conditions: skin and soft tissue infection, pneumonia, septicemia, peritonitis, bacteremia, and endocarditis. Mice have been used extensively in all these conditions to identify virulence factors and host components important for pathogenesis. Although significant effort has gone toward development of an anti-staphylococcal vaccine, antibodies have proven ineffective in preventing infection in humans after successful studies in mice. These results have raised questions as to the utility of mice to predict patient outcome and suggest that humanized mice might prove useful in modeling infection. The development of humanized mouse models of S. aureus infection will allow us to assess the contribution of several human-specific virulence factors, in addition to exploring components of the human immune system in protection against S. aureus infection. Their use is discussed in light of several recently reported studies.
Collapse
Affiliation(s)
- Dane Parker
- Department of Pediatrics, Columbia University, New York, NY, USA
| |
Collapse
|
22
|
Towards a Humanized Mouse Model of Liver Stage Malaria Using Ectopic Artificial Livers. Sci Rep 2017; 7:45424. [PMID: 28361899 PMCID: PMC5374446 DOI: 10.1038/srep45424] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/01/2017] [Indexed: 12/11/2022] Open
Abstract
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.
Collapse
|
23
|
Winer BY, Huang T, Low BE, Avery C, Pais MA, Hrebikova G, Siu E, Chiriboga L, Wiles MV, Ploss A. Recapitulation of treatment response patterns in a novel humanized mouse model for chronic hepatitis B virus infection. Virology 2016; 502:63-72. [PMID: 28006671 DOI: 10.1016/j.virol.2016.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/12/2016] [Accepted: 12/14/2016] [Indexed: 02/08/2023]
Abstract
There are ~350 million chronic carriers of hepatitis B (HBV). While a prophylactic vaccine and drug regimens to suppress viremia are available, chronic HBV infection is rarely cured. HBV's limited host tropism leads to a scarcity of susceptible small animal models and is a hurdle to developing curative therapies. Mice that support engraftment with human hepatoctyes have traditionally been generated through crosses of murine liver injury models to immunodeficient backgrounds. Here, we describe the disruption of fumarylacetoacetate hydrolase directly in the NOD Rag1-/- IL2RγNULL (NRG) background using zinc finger nucleases. The resultant human liver chimeric mice sustain persistent HBV viremia for >90 days. When treated with standard of care therapy, HBV DNA levels decrease below detection but rebound when drug suppression is released, mimicking treatment response observed in patients. Our study highlights the utility of directed gene targeting approaches in zygotes to create new humanized mouse models for human diseases.
Collapse
Affiliation(s)
- Benjamin Y Winer
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA
| | - Tiffany Huang
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA
| | - Benjamin E Low
- Department of Technology Evaluation and Development, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609-1500 USA
| | - Cindy Avery
- Department of Technology Evaluation and Development, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609-1500 USA
| | - Mihai-Alexandru Pais
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA
| | - Gabriela Hrebikova
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA
| | - Evelyn Siu
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA
| | - Luis Chiriboga
- Department of Pathology, New York University Medical Center, New York, NY 10016, USA
| | - Michael V Wiles
- Department of Technology Evaluation and Development, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609-1500 USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey, NJ 08544, USA.
| |
Collapse
|
24
|
Raphemot R, Posfai D, Derbyshire ER. Current therapies and future possibilities for drug development against liver-stage malaria. J Clin Invest 2016; 126:2013-20. [PMID: 27249674 DOI: 10.1172/jci82981] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Malaria remains a global public health threat, with half of the world's population at risk. Despite numerous efforts in the past decade to develop new antimalarial drugs to surmount increasing resistance to common therapies, challenges remain in the expansion of the current antimalarial arsenal for the elimination of this disease. The requirement of prophylactic and radical cure activities for the next generation of antimalarial drugs demands that new research models be developed to support the investigation of the elusive liver stage of the malaria parasite. In this Review, we revisit current antimalarial therapies and discuss recent advances for in vitro and in vivo malaria research models of the liver stage and their importance in probing parasite biology and the discovery of novel drug candidates.
Collapse
|
25
|
Hentzschel F, Herrmann AK, Mueller AK, Grimm D. Plasmodium meets AAV-the (un)likely marriage of parasitology and virology, and how to make the match. FEBS Lett 2016; 590:2027-45. [PMID: 27117587 DOI: 10.1002/1873-3468.12187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 03/24/2016] [Accepted: 04/21/2016] [Indexed: 01/21/2023]
Abstract
The increasing use of screening technologies in malaria research has substantially expanded our knowledge on cellular factors hijacked by the Plasmodium parasite in the infected host, including those that participate in the clinically silent liver stage. This rapid gain in our understanding of the hepatic interaction partners now requires a means to validate and further disentangle parasite-host networks in physiologically relevant liver model systems. Here, we outline seminal work that contributed to our present knowledge on the intrahepatic Plasmodium host factors, followed by a discussion of surrogate models of mammalian livers or hepatocytes. We finally describe how Adeno-associated viruses could be engineered and used as hepatotropic tools to dissect Plasmodium-host interactions, and to deliberately control these networks for antimalaria vaccination or therapy.
Collapse
Affiliation(s)
- Franziska Hentzschel
- Department of Parasitology, Center for Infectious Diseases, Heidelberg University Hospital, Germany.,Department of Virology, Center for Infectious Diseases, Heidelberg University Hospital, Germany.,Cluster of Excellence CellNetworks, Heidelberg, Germany
| | - Anne-Kathrin Herrmann
- Department of Virology, Center for Infectious Diseases, Heidelberg University Hospital, Germany.,Cluster of Excellence CellNetworks, Heidelberg, Germany
| | - Ann-Kristin Mueller
- Department of Parasitology, Center for Infectious Diseases, Heidelberg University Hospital, Germany
| | - Dirk Grimm
- Department of Virology, Center for Infectious Diseases, Heidelberg University Hospital, Germany.,Cluster of Excellence CellNetworks, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| |
Collapse
|
26
|
Noulin F. Malaria modeling: In vitro stem cells vs in vivo models. World J Stem Cells 2016; 8:88-100. [PMID: 27022439 PMCID: PMC4807312 DOI: 10.4252/wjsc.v8.i3.88] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/07/2015] [Accepted: 01/29/2016] [Indexed: 02/06/2023] Open
Abstract
The recent development of stem cell research and the possibility of generating cells that can be stably and permanently modified in their genome open a broad horizon in the world of in vitro modeling. The malaria field is gaining new opportunities from this important breakthrough and novel tools were adapted and opened new frontiers for malaria research. In addition to the new in vitro systems, in recent years there were also significant advances in the development of new animal models that allows studying the entire cell cycle of human malaria. In this paper, we review the different protocols available to study human Plasmodium species either by using stem cell or alternative animal models.
Collapse
|
27
|
Meier RPH, Navarro-Alvarez N, Morel P, Schuurman HJ, Strom S, Bühler LH. Current status of hepatocyte xenotransplantation. Int J Surg 2015; 23:273-279. [PMID: 26361861 DOI: 10.1016/j.ijsu.2015.08.077] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 07/29/2015] [Accepted: 08/05/2015] [Indexed: 12/14/2022]
Abstract
The treatment of acute liver failure, a condition with high mortality, comprises optimal clinical care, and in severe cases liver transplantation. However, there are limitations in availability of organ donors. Hepatocyte transplantation is a promising alternative that could fill the medical need, in particular as the bridge to liver transplantation. Encapsulated porcine hepatocytes represent an unlimited source that could function as a bioreactor requiring minimal immunosuppression. Besides patients with acute liver failure, patients with alcoholic hepatitis who are unresponsive to a short course of corticosteroids are a target for hepatocyte transplantation. In this review we present an overview of the innate immune barriers in hepatocyte xenotransplantation, including the role of complement and natural antibodies; the role of phagocytic cells and ligands like CD47 in the regulation of phagocytic cells; and the role of Natural Killer cells. We present also some illustrations of physiological species incompatibilities in hepatocyte xenotransplantation, such as incompatibilities in the coagulation system. An overview of the methodology for cell microencapsulation is presented, followed by proof-of-concept studies in rodent and nonhuman primate models of fulminant liver failure: these studies document the efficacy of microencapsulated porcine hepatocytes which warrants progress towards clinical application. Lastly, we present an outline of a provisional clinical trial, that upon completion of preclinical work could start within the upcoming 2-3 years.
Collapse
Affiliation(s)
- Raphael P H Meier
- Visceral and Transplantation Surgery, Department of Surgery, University Hospitals of Geneva and Faculty of Medicine, Geneva, Switzerland.
| | - Nalu Navarro-Alvarez
- Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Philippe Morel
- Visceral and Transplantation Surgery, Department of Surgery, University Hospitals of Geneva and Faculty of Medicine, Geneva, Switzerland
| | - Henk-Jan Schuurman
- Visceral and Transplantation Surgery, Department of Surgery, University Hospitals of Geneva and Faculty of Medicine, Geneva, Switzerland
| | - Stephen Strom
- Cell Transplantation and Regenerative Medicine, Department of Laboratory Medicine, Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Leo H Bühler
- Visceral and Transplantation Surgery, Department of Surgery, University Hospitals of Geneva and Faculty of Medicine, Geneva, Switzerland
| |
Collapse
|
28
|
Humanized Mouse Models to Study Cell-Mediated Immune Responses to Liver-Stage Malaria Vaccines. Trends Parasitol 2015; 31:583-594. [PMID: 26458783 DOI: 10.1016/j.pt.2015.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 12/23/2022]
Abstract
Malaria vaccine development is hampered by the lack of small animal models that recapitulate human immune responses to Plasmodium falciparum. We review the burgeoning literature on humanized mice for P. falciparum infection, including challenges in engraftment of human immune cells, hepatocytes, and erythrocytes. Recent advances in immune-compromised mouse models and stem cell technology have already enabled proof of concept that the entire parasite life cycle can be sustained in a murine model and that adaptive human immune responses to several parasite stages can be measured. Nonetheless, optimization is needed to achieve a reproducible and relevant murine model for malaria vaccine development. This review is focused on the complexities of T cell development in a mouse humanized with both a lymphoid system and hepatocytes. An understanding of this will facilitate the use of humanized mice in the development of liver-stage vaccines.
Collapse
|
29
|
Longley RJ, Hill AVS, Spencer AJ. Malaria vaccines: identifying Plasmodium falciparum liver-stage targets. Front Microbiol 2015; 6:965. [PMID: 26441899 PMCID: PMC4569888 DOI: 10.3389/fmicb.2015.00965] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 08/31/2015] [Indexed: 01/08/2023] Open
Abstract
The development of a highly efficacious and durable vaccine for malaria remains a top priority for global health researchers. Despite the huge rise in recognition of malaria as a global health problem and the concurrent rise in funding over the past 10–15 years, malaria continues to remain a widespread burden. The evidence of increasing resistance to anti-malarial drugs and insecticides is a growing concern. Hence, an efficacious and durable preventative vaccine for malaria is urgently needed. Vaccines are one of the most cost-effective tools and have successfully been used in the prevention and control of many diseases, however, the development of a vaccine for the Plasmodium parasite has proved difficult. Given the early success of whole sporozoite mosquito-bite delivered vaccination strategies, we know that a vaccine for malaria is an achievable goal, with sub-unit vaccines holding great promise as they are simple and cheap to both manufacture and deploy. However a major difficulty in development of sub-unit vaccines lies within choosing the appropriate antigenic target from the 5000 or so genes expressed by the parasite. Given the liver-stage of malaria represents a bottle-neck in the parasite’s life cycle, there is widespread agreement that a multi-component sub-unit malaria vaccine should preferably contain a liver-stage target. In this article we review progress in identifying and screening Plasmodium falciparum liver-stage targets for use in a malaria vaccine.
Collapse
Affiliation(s)
- Rhea J Longley
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford Oxford, UK
| | - Adrian V S Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford Oxford, UK
| | - Alexandra J Spencer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford Oxford, UK
| |
Collapse
|
30
|
Soulard V, Bosson-Vanga H, Lorthiois A, Roucher C, Franetich JF, Zanghi G, Bordessoulles M, Tefit M, Thellier M, Morosan S, Le Naour G, Capron F, Suemizu H, Snounou G, Moreno-Sabater A, Mazier D. Plasmodium falciparum full life cycle and Plasmodium ovale liver stages in humanized mice. Nat Commun 2015. [PMID: 26205537 PMCID: PMC4525212 DOI: 10.1038/ncomms8690] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Experimental studies of Plasmodium parasites that infect humans are restricted by their host specificity. Humanized mice offer a means to overcome this and further provide the opportunity to observe the parasites in vivo. Here we improve on previous protocols to achieve efficient double engraftment of TK-NOG mice by human primary hepatocytes and red blood cells. Thus, we obtain the complete hepatic development of P. falciparum, the transition to the erythrocytic stages, their subsequent multiplication, and the appearance of mature gametocytes over an extended period of observation. Furthermore, using sporozoites derived from two P. ovale-infected patients, we show that human hepatocytes engrafted in TK-NOG mice sustain maturation of the liver stages, and the presence of late-developing schizonts indicate the eventual activation of quiescent parasites. Thus, TK-NOG mice are highly suited for in vivo observations on the Plasmodium species of humans. Mice engrafted with human cells are useful models for research on human malaria parasites. Here the authors show that the complete life cycle of Plasmodium falciparum and the liver stages of Plasmodium ovale can be studied in mice doubly engrafted with human primary hepatocytes and red blood cells.
Collapse
Affiliation(s)
- Valérie Soulard
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France
| | - Henriette Bosson-Vanga
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [4] Université FHB, UFR SPB, Departement de Parasitologie-Mycologie, BP V 34 Abidjan, Ivory Coast
| | - Audrey Lorthiois
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France
| | - Clémentine Roucher
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France
| | - Jean-François Franetich
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France
| | - Gigliola Zanghi
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France
| | - Mallaury Bordessoulles
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France
| | - Maurel Tefit
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France
| | - Marc Thellier
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service Parasitologie-Mycologie, Centre National de Référence du Paludisme, 83 Bd de l'hôpital, F-75013 Paris, France
| | - Serban Morosan
- UPMC Univ. Paris 06, INSERM, UMS28, 105 Bd de l'hôpital, F-75013 Paris, France
| | - Gilles Le Naour
- AP-HP, UPMC Univ. Paris 06, Groupe Hospitalier Pitié-Salpêtrière, Service d'anatomie et cytologie pathologiques, 83 Bd de l'hôpital, F-75013 Paris, France
| | - Frédérique Capron
- AP-HP, UPMC Univ. Paris 06, Groupe Hospitalier Pitié-Salpêtrière, Service d'anatomie et cytologie pathologiques, 83 Bd de l'hôpital, F-75013 Paris, France
| | - Hiroshi Suemizu
- Central Institute for Experimental Animal, Kawasaki, Kanegawa, Japan
| | - Georges Snounou
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France
| | - Alicia Moreno-Sabater
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France
| | - Dominique Mazier
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 91 Bd de l'hôpital, F-75013 Paris, France [2] INSERM, U1135, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [3] CNRS, ERL 8255, CIMI-PARIS, 91 Bd de l'hôpital, F-75013 Paris, France [4] AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service Parasitologie-Mycologie, Centre National de Référence du Paludisme, 83 Bd de l'hôpital, F-75013 Paris, France
| |
Collapse
|
31
|
Kaushansky A, Kappe SH. Selection and refinement: the malaria parasite's infection and exploitation of host hepatocytes. Curr Opin Microbiol 2015; 26:71-8. [PMID: 26102161 DOI: 10.1016/j.mib.2015.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/16/2015] [Accepted: 05/22/2015] [Indexed: 01/23/2023]
Abstract
Plasmodium parasites belong to the Apicomplexan phylum, which consists mostly of obligate intracellular pathogens that vary dramatically in host cell tropism. Plasmodium sporozoites are highly hepatophilic. The specific molecular mechanisms, which facilitate sporozoite selection and successful infection of hepatocytes, remain poorly defined. Here, we discuss the parasite and host factors which are critical to hepatocyte infection. We derive a model where sporozoites initially select host cells that constitute a permissive environment and then further refine the chosen hepatocyte during liver stage development, ensuring life cycle progression. While many unknowns of pre-erythrocytic infection remain, advancing models and technologies that enable analysis of human malaria parasites and of single infected cells will catalyze a comprehensive understanding of the interaction between the malaria parasite and its hepatocyte host.
Collapse
Affiliation(s)
- Alexis Kaushansky
- Center for Infectious Disease Research, Formerly Seattle Biomedical Research Institute, 307 Westlake Ave. N, #500, Seattle, WA 98109, United States.
| | - Stefan Hi Kappe
- Center for Infectious Disease Research, Formerly Seattle Biomedical Research Institute, 307 Westlake Ave. N, #500, Seattle, WA 98109, United States; Department of Global Health, University of Washington, Seattle, WA, United States.
| |
Collapse
|
32
|
Lin C, Ballinger KR, Khetani SR. The application of engineered liver tissues for novel drug discovery. Expert Opin Drug Discov 2015; 10:519-40. [PMID: 25840592 DOI: 10.1517/17460441.2015.1032241] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Drug-induced liver injury remains a major cause of drug attrition. Furthermore, novel drugs are being developed for treating liver diseases. However, differences between animals and humans in liver pathways necessitate the use of human-relevant liver models to complement live animal testing during preclinical drug development. Microfabrication tools and synthetic biomaterials now allow for the creation of tissue subunits that display more physiologically relevant and long-term liver functions than possible with declining monolayers. AREAS COVERED The authors discuss acellular enzyme platforms, two-dimensional micropatterned co-cultures, three-dimensional spheroidal cultures, microfluidic perfusion, liver slices and humanized rodent models. They also present the use of cell lines, primary liver cells and induced pluripotent stem cell-derived human hepatocyte-like cells in the creation of cell-based models and discuss in silico approaches that allow integration and modeling of the datasets from these models. Finally, the authors describe the application of liver models for the discovery of novel therapeutics for liver diseases. EXPERT OPINION Engineered liver models with varying levels of in vivo-like complexities provide investigators with the opportunity to develop assays with sufficient complexity and required throughput. Control over cell-cell interactions and co-culture with stromal cells in both two dimension and three dimension are critical for enabling stable liver models. The validation of liver models with diverse sets of compounds for different applications, coupled with an analysis of cost:benefit ratio, is important for model adoption for routine screening. Ultimately, engineered liver models could significantly reduce drug development costs and enable the development of more efficacious and safer therapeutics for liver diseases.
Collapse
Affiliation(s)
- Christine Lin
- Colorado State University, School of Biomedical Engineering , 200 W. Lake St, 1301 Campus Delivery, Fort Collins, CO 80523-1374 , USA
| | | | | |
Collapse
|
33
|
Strick-Marchand H, Dusséaux M, Darche S, Huntington ND, Legrand N, Masse-Ranson G, Corcuff E, Ahodantin J, Weijer K, Spits H, Kremsdorf D, Di Santo JP. A novel mouse model for stable engraftment of a human immune system and human hepatocytes. PLoS One 2015; 10:e0119820. [PMID: 25782010 PMCID: PMC4364106 DOI: 10.1371/journal.pone.0119820] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/16/2015] [Indexed: 01/27/2023] Open
Abstract
Hepatic infections by hepatitis B virus (HBV), hepatitis C virus (HCV) and Plasmodium parasites leading to acute or chronic diseases constitute a global health challenge. The species tropism of these hepatotropic pathogens is restricted to chimpanzees and humans, thus model systems to study their pathological mechanisms are severely limited. Although these pathogens infect hepatocytes, disease pathology is intimately related to the degree and quality of the immune response. As a first step to decipher the immune response to infected hepatocytes, we developed an animal model harboring both a human immune system (HIS) and human hepatocytes (HUHEP) in BALB/c Rag2-/- IL-2Rγc-/- NOD.sirpa uPAtg/tg mice. The extent and kinetics of human hepatocyte engraftment were similar between HUHEP and HIS-HUHEP mice. Transplanted human hepatocytes were polarized and mature in vivo, resulting in 20-50% liver chimerism in these models. Human myeloid and lymphoid cell lineages developed at similar frequencies in HIS and HIS-HUHEP mice, and splenic and hepatic compartments were humanized with mature B cells, NK cells and naïve T cells, as well as monocytes and dendritic cells. Taken together, these results demonstrate that HIS-HUHEP mice can be stably (> 5 months) and robustly engrafted with a humanized immune system and chimeric human liver. This novel HIS-HUHEP model provides a platform to investigate human immune responses against hepatotropic pathogens and to test novel drug strategies or vaccine candidates.
Collapse
Affiliation(s)
- Helene Strick-Marchand
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U668, Paris, France
| | - Mathilde Dusséaux
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U668, Paris, France
| | - Sylvie Darche
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U668, Paris, France
| | - Nicholas D. Huntington
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U668, Paris, France
| | - Nicolas Legrand
- Academic Medical Center at the University of Amsterdam, Amsterdam, The Netherlands
| | - Guillemette Masse-Ranson
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U668, Paris, France
| | - Erwan Corcuff
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U668, Paris, France
| | - James Ahodantin
- Institut National de la Santé et de la Recherche Médicale (INSERM) U845, Faculté de Médecine Paris Descartes, Paris, France
| | - Kees Weijer
- Academic Medical Center at the University of Amsterdam, Amsterdam, The Netherlands
| | - Hergen Spits
- Academic Medical Center at the University of Amsterdam, Amsterdam, The Netherlands
| | - Dina Kremsdorf
- Institut National de la Santé et de la Recherche Médicale (INSERM) U845, Faculté de Médecine Paris Descartes, Paris, France
| | - James P. Di Santo
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U668, Paris, France
- * E-mail:
| |
Collapse
|
34
|
Matsumoto Y, Ishii M, Ishii K, Miyaguchi W, Horie R, Inagaki Y, Hamamoto H, Tatematsu KI, Uchino K, Tamura T, Sezutsu H, Sekimizu K. Transgenic silkworms expressing human insulin receptors for evaluation of therapeutically active insulin receptor agonists. Biochem Biophys Res Commun 2014; 455:159-64. [DOI: 10.1016/j.bbrc.2014.10.143] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/28/2014] [Indexed: 11/29/2022]
|
35
|
Wijayalath W, Majji S, Villasante EF, Brumeanu TD, Richie TL, Casares S. Humanized HLA-DR4.RagKO.IL2RγcKO.NOD (DRAG) mice sustain the complex vertebrate life cycle of Plasmodium falciparum malaria. Malar J 2014; 13:386. [PMID: 25266106 PMCID: PMC4197321 DOI: 10.1186/1475-2875-13-386] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/18/2014] [Indexed: 12/28/2022] Open
Abstract
Background Malaria is a deadly infectious disease affecting millions of people in tropical and sub-tropical countries. Among the five species of Plasmodium parasites that infect humans, Plasmodium falciparum accounts for the highest morbidity and mortality associated with malaria. Since humans are the only natural hosts for P. falciparum, the lack of convenient animal models has hindered the understanding of disease pathogenesis and prompted the need of testing anti-malarial drugs and vaccines directly in human trials. Humanized mice hosting human cells represent new pre-clinical models for infectious diseases that affect only humans. In this study, the ability of human-immune-system humanized HLA-DR4.RagKO.IL2RγcKO.NOD (DRAG) mice to sustain infection with P. falciparum was explored. Methods Four week-old DRAG mice were infused with HLA-matched human haematopoietic stem cells (HSC) and examined for reconstitution of human liver cells and erythrocytes. Upon challenge with infectious P. falciparum sporozoites (NF54 strain) humanized DRAG mice were examined for liver stage infection, blood stage infection, and transmission to Anopheles stephensi mosquitoes. Results Humanized DRAG mice reconstituted human hepatocytes, Kupffer cells, liver endothelial cells, and erythrocytes. Upon intravenous challenge with P. falciparum sporozoites, DRAG mice sustained liver to blood stage infection (average 3–5 parasites/microlitre blood) and allowed transmission to An. stephensi mosquitoes. Infected DRAG mice elicited antibody and cellular responses to the blood stage parasites and self-cured the infection by day 45 post-challenge. Conclusions DRAG mice represent the first human-immune-system humanized mouse model that sustains the complex vertebrate life cycle of P. falciparum without the need of exogenous injection of human hepatocytes/erythrocytes or P. falciparum parasite adaptation. The ability of DRAG mice to elicit specific human immune responses to P. falciparum parasites may help deciphering immune correlates of protection and to identify protective malaria antigens.
Collapse
Affiliation(s)
| | | | | | | | | | - Sofia Casares
- US Military Malaria Vaccine Program, Naval Medical Research Center/Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA.
| |
Collapse
|
36
|
Wilson EM, Bial J, Tarlow B, Bial G, Jensen B, Greiner DL, Brehm MA, Grompe M. Extensive double humanization of both liver and hematopoiesis in FRGN mice. Stem Cell Res 2014; 13:404-12. [PMID: 25310256 PMCID: PMC7275629 DOI: 10.1016/j.scr.2014.08.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 08/21/2014] [Accepted: 08/27/2014] [Indexed: 11/21/2022] Open
Abstract
Preclinical research in animals often fails to adequately predict the outcomes observed in human patients. Chimeric animals bearing individual human tissues have been developed to provide improved models of human-specific cellular processes. Mice transplanted with human hematopoietic stem cells can be used to study human immune responses, infections of blood cells and processes of hematopoiesis. Animals with humanized livers are useful for modeling hepatotropic infections as well as drug metabolism and hepatotoxicity. However, many pathophysiologic processes involve both the liver and the hematolymphoid system. Examples include hepatitis C/HIV co-infection, immune mediated liver diseases, liver injuries with inflammation such as steatohepatitis and alcoholic liver disease. We developed a robust protocol enabling the concurrent double-humanization of mice with mature hepatocytes and human blood. Immune-deficient, fumarylacetoacetate hydrolase (Fah−/−), Rag2−/− and Il2rg−/− deficient animals on the NOD-strain background (FRGN) were simultaneously co-transplanted with adult human hepatocytes and hematopoietic stem cells after busulfan and Ad:uPA pre-conditioning. Four months after transplantation the average human liver repopulation exceeded 80% and hematopoietic chimerism also was high (40–80% in bone marrow). Importantly, human macrophages (Kupffer cells) were present in the chimeric livers. Double-chimeric FRGN mice will serve as a new model for disease processes that involve interactions between hepatocytes and hematolymphoid cells.
Collapse
Affiliation(s)
| | - J Bial
- Yecuris Corp., Tigard, OR, USA
| | | | - G Bial
- Yecuris Corp., Tigard, OR, USA
| | | | - D L Greiner
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - M A Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | |
Collapse
|
37
|
Brehm MA, Wiles MV, Greiner DL, Shultz LD. Generation of improved humanized mouse models for human infectious diseases. J Immunol Methods 2014; 410:3-17. [PMID: 24607601 PMCID: PMC4155027 DOI: 10.1016/j.jim.2014.02.011] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 02/18/2014] [Indexed: 12/26/2022]
Abstract
The study of human-specific infectious agents has been hindered by the lack of optimal small animal models. More recently development of novel strains of immunodeficient mice has begun to provide the opportunity to utilize small animal models for the study of many human-specific infectious agents. The introduction of a targeted mutation in the IL2 receptor common gamma chain gene (IL2rg(null)) in mice already deficient in T and B cells led to a breakthrough in the ability to engraft hematopoietic stem cells, as well as functional human lymphoid cells and tissues, effectively creating human immune systems in immunodeficient mice. These humanized mice are becoming increasingly important as pre-clinical models for the study of human immunodeficiency virus-1 (HIV-1) and other human-specific infectious agents. However, there remain a number of opportunities to further improve humanized mouse models for the study of human-specific infectious agents. This is being done by the implementation of innovative technologies, which collectively will accelerate the development of new models of genetically modified mice, including; i) modifications of the host to reduce innate immunity, which impedes human cell engraftment; ii) genetic modification to provide human-specific growth factors and cytokines required for optimal human cell growth and function; iii) and new cell and tissue engraftment protocols. The development of "next generation" humanized mouse models continues to provide exciting opportunities for the establishment of robust small animal models to study the pathogenesis of human-specific infectious agents, as well as for testing the efficacy of therapeutic agents and experimental vaccines.
Collapse
Affiliation(s)
- Michael A Brehm
- The University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, United States.
| | - Michael V Wiles
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, United States.
| | - Dale L Greiner
- The University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, United States.
| | - Leonard D Shultz
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, United States.
| |
Collapse
|
38
|
Zuzarte-Luis V, Mota MM, Vigário AM. Malaria infections: what and how can mice teach us. J Immunol Methods 2014; 410:113-22. [PMID: 24837740 DOI: 10.1016/j.jim.2014.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/24/2014] [Accepted: 05/01/2014] [Indexed: 01/07/2023]
Abstract
Malaria imposes a horrific public health burden - hundreds of millions of infections and millions of deaths - on large parts of the world. While this unacceptable health burden and its economic and social impact have made it a focal point of the international development agenda, it became consensual that malaria control or elimination will be difficult to attain prior to gain a better understanding of the complex interactions occurring between its main players: Plasmodium, the causative agent of disease, and its hosts. Practical and ethical limitations exist regarding the ability to carry out research with human subjects or with human samples. In this review, we highlight how rodent models of infection have contributed significantly during the past decades to a better understanding of the basic biology of the parasite, host response and pathogenesis.
Collapse
Affiliation(s)
- Vanessa Zuzarte-Luis
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Maria M Mota
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Ana M Vigário
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal; Unidade de Ciências Médicas, Centro de Competência de Ciências da Vida, Universidade da Madeira, Funchal, Portugal.
| |
Collapse
|
39
|
Angulo-Barturen I, Ferrer S. Humanised models of infection in the evaluation of anti-malarial drugs. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 10:e351-7. [PMID: 24050131 DOI: 10.1016/j.ddtec.2012.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Humanised mice have a crucial role for drug discovery in malaria, which is the most important parasitic disease in the world and is caused by protozoa of the genus Plasmodium that selectively infect human hepatocytes and erythrocytes. There are currently reliable humanised murine models for hepatic and erythrocytic stages of Plasmodium falciparum, which is the most pathogenic malarial species. These models are useful in the evaluation of drugs for malaria prevention and treatment, notably in exploiting the thousands of antimalarial hits discovered. The development of a humanised model for Plasmodium vivax and the validation of the P. falciparum models to inform optimal clinical studies are the next key goals to be achieved.
Collapse
|
40
|
Foquet L, Hermsen CC, van Gemert GJ, Van Braeckel E, Weening KE, Sauerwein R, Meuleman P, Leroux-Roels G. Vaccine-induced monoclonal antibodies targeting circumsporozoite protein prevent Plasmodium falciparum infection. J Clin Invest 2014; 124:140-4. [PMID: 24292709 DOI: 10.1172/jci70349] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 09/26/2013] [Indexed: 11/17/2022] Open
Abstract
Malaria, which is the result of Plasmodium falciparum infection, is a global health threat that resulted in 655,000 deaths and 216 million clinical cases in 2010 alone. Recent phase 3 trials with malaria vaccine candidate RTS,S/AS01 (RTS,S) in children has demonstrated modest efficacy against clinical and severe malaria. RTS,S targets the pre-erythrocytic phase of the disease and induces high antibody titers against the P. falciparum circumsporozoite protein (CSP) and a moderate CD4(+) T cell response. The individual contribution of these adaptive immune responses to protection from infection remains unknown. Here, we found that prophylactic administration of anti-CSP mAbs derived from an RTS,S-vaccinated recipient fully protected mice with humanized livers from i.v.- and mosquito bite–delivered P. falciparum sporozoite challenge. Titers of anti-CSP that conveyed full protection were within the range observed in human RTS,S vaccine recipients. Increasing anti-CSP titers resulted in a dose-dependent reduction of the liver parasite burden. These data indicate that RTS,S-induced antibodies are protective and provide sterilizing immunity against P. falciparum infection when reaching or exceeding a critical plasma concentration.
Collapse
|
41
|
Foster JR, Lund G, Sapelnikova S, Tyrrell DL, Kneteman NM. Chimeric rodents with humanized liver: bridging the preclinical/clinical trial gap in ADME/toxicity studies. Xenobiotica 2013; 44:109-22. [DOI: 10.3109/00498254.2013.867553] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
42
|
Grompe M, Strom S. Mice with human livers. Gastroenterology 2013; 145:1209-14. [PMID: 24042096 DOI: 10.1053/j.gastro.2013.09.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 09/04/2013] [Accepted: 09/04/2013] [Indexed: 12/28/2022]
Abstract
Animal models are used to study many aspects of human disease and to test therapeutic interventions. However, some very important features of human biology cannot be replicated in animals, even in nonhuman primates or transgenic rodents engineered with human genes. Most human microbial pathogens do not infect animals and the metabolism of many xenobiotics is different between human beings and animals. The advent of transgenic immune-deficient mice has made it possible to generate chimeric animals harboring human tissues and cells, including hepatocytes. The liver plays a central role in many human-specific biological processes and mice with humanized livers can be used to model human metabolism, liver injury, gene regulation, drug toxicity, and hepatotropic infections.
Collapse
Affiliation(s)
- Markus Grompe
- Oregon Stem Cell Center, Department of Pediatrics, Oregon Health & Science University, Portland, Oregon.
| | | |
Collapse
|
43
|
Melican K, Duménil G. A humanized model of microvascular infection. Future Microbiol 2013; 8:567-9. [PMID: 23642111 DOI: 10.2217/fmb.13.35] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
44
|
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: 1.5] [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
|
45
|
Abstract
The emergence of resistance to artemisinins and the renewed efforts to eradicate malaria demand the urgent development of new drugs. In this endeavour, the evaluation of efficacy in animal models is often a go/no go decision assay in drug discovery. This important role relies on the capability of animal models to assess the disposition, toxicology and efficacy of drugs in a single test. Although the relative merits of each efficacy model of malaria as human surrogate have been extensively discussed, there are no critical analyses on the use of such models in current drug discovery. In this article, we intend to analyse how efficacy models are used to discover new antimalarial drugs. Our analysis indicates that testing drug efficacy is often the last assay in each discovery stage and the experimental designs utilized are not optimized to expedite decision-making and inform clinical development. In light of this analysis, we propose new ways to accelerate drug discovery using efficacy models.
Collapse
|
46
|
Brehm MA, Jouvet N, Greiner DL, Shultz LD. Humanized mice for the study of infectious diseases. Curr Opin Immunol 2013; 25:428-35. [PMID: 23751490 DOI: 10.1016/j.coi.2013.05.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/15/2013] [Accepted: 05/17/2013] [Indexed: 12/17/2022]
Abstract
Many of the pathogens that cause human infectious diseases do not infect rodents or other mammalian species. Small animal models that allow studies of the pathogenesis of these agents and evaluation of drug efficacy are critical for identifying ways to prevent and treat human infectious diseases. Immunodeficient mice engrafted with functional human cells and tissues, termed 'humanized' mice, represent a critical pre-clinical bridge for in vivo studies of human pathogens. Recent advances in the development of humanized mice have allowed in vivo studies of multiple human infectious agents providing novel insights into their pathogenesis that was otherwise not possible.
Collapse
Affiliation(s)
- Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, United States
| | | | | | | |
Collapse
|
47
|
Kawahara T, Lisboa LF, Cader S, Douglas DN, Nourbakhsh M, Pu CH, Lewis JT, Churchill TA, Humar A, Kneteman NM. Human cytomegalovirus infection in humanized liver chimeric mice. Hepatol Res 2013; 43:679-84. [PMID: 23442000 DOI: 10.1111/j.1872-034x.2012.01116.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 10/02/2012] [Accepted: 10/04/2012] [Indexed: 02/08/2023]
Abstract
AIM Cytomegalovirus is a common viral pathogen that influences the outcome of organ transplantation. To date, there is no established method to evaluate the effects of human CMV (HCMV) treatments in vivo except for human clinical trials. In the current study, we describe the development of a mouse model that supports the in vivo propagation of HCMV. METHODS One million viable human hepatocytes, purified from human livers, were injected into the spleens of severe combined immunodeficient/albumin linked-urokinase type plasminogen activator transgenic mice. A clinical strain of HCMV was inoculated in mice with confirmed human hepatocyte engraftment or in non-chimeric controls. Infection was monitored through HCMV titers in the plasma. Mice were administrated ganciclovir (50 mg/kg per day, i.p.) beginning at 2 days post-HCMV inoculation, or human liver natural killer (NK) cells (20 × 10(6) cells/mouse, i.v.) 1 day prior to HCMV inoculation. RESULTS Chimeric mice that received HCMV showed high plasma titers of HCMV DNA on days 1 and 6 that became undetectable by day 11 post-inoculation. In contrast, non-transplanted mice had only residual plasma inoculum detection at day 1 and no detectable viremia thereafter. The levels of HCMV DNA were reduced by ganciclovir treatment or by human liver NK cell adoptive transfer, while HCMV-infected chimeric mice that were not treated sustained viremia during the follow up. CONCLUSION Human liver chimeric mice provide an in vivo model for the study of acute HCMV infection of hepatocytes.
Collapse
Affiliation(s)
- Toshiyasu Kawahara
- Division of Transplantation Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Liu Y, Meyer C, Xu C, Weng H, Hellerbrand C, ten Dijke P, Dooley S. Animal models of chronic liver diseases. Am J Physiol Gastrointest Liver Physiol 2013; 304:G449-G468. [PMID: 23275613 DOI: 10.1152/ajpgi.00199.2012] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Chronic liver diseases are frequent and potentially life threatening for humans. The underlying etiologies are diverse, ranging from viral infections, autoimmune disorders, and intoxications (including alcohol abuse) to imbalanced diets. Although at early stages of disease the liver regenerates in the absence of the insult, advanced stages cannot be healed and may require organ transplantation. A better understanding of underlying mechanisms is mandatory for the design of new drugs to be used in clinic. Therefore, rodent models are being developed to mimic human liver disease. However, no model to date can completely recapitulate the "corresponding" human disorder. Limiting factors are the time frame required in humans to establish a certain liver disease and the fact that rodents possess a distinct immune system compared with humans and have different metabolic rates affecting liver homeostasis. These features account for the difficulties in developing adequate rodent models for studying disease progression and for testing new pharmaceuticals to be translated into the clinic. Nevertheless, traditional and new promising animal models that mimic certain attributes of chronic liver diseases are established and being used to deepen our understanding in the underlying mechanisms of distinct liver diseases. This review aims at providing a comprehensive overview of recent advances in animal models recapitulating different features and etiologies of human liver diseases.
Collapse
Affiliation(s)
- Yan Liu
- Department of Medicine II, Section Molecular Hepatology-Alcohol Associated Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | | | | | | | | |
Collapse
|
49
|
Waern JM, Yuan Q, Rüdrich U, Becker PD, Schulze K, Strick-Marchand H, Huntington ND, Zacher BJ, Wursthorn K, DiSanto JP, Guzman CA, Manns MP, Ott M, Bock M. Ectopic expression of murine CD47 minimizes macrophage rejection of human hepatocyte xenografts in immunodeficient mice. Hepatology 2012; 56:1479-88. [PMID: 22535707 DOI: 10.1002/hep.25816] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Macrophages play an important role in the rejection of xenogeneic cells and therefore represent a major obstacle to generating chimeric mice with human xenografts that are useful tools for basic and preclinical medical research. The signal inhibitory regulatory protein α (SIRPα) receptor is a negative regulator of macrophage phagocytic activity and interacts in a species-specific fashion with its ligand CD47. Furthermore, SIRPα polymorphism in laboratory mouse strains significantly affects the extent of human CD47-mediated toleration of human xenotransplants. Aiming to minimize macrophage activity and thus optimize human cell engraftment in immunodeficient mice, we lentivirally transduced murine CD47 (Cd47) into human liver cells. Human HepG2 liver cells expressing Cd47 were less frequently contacted and phagocytosed by murine RAW264.7 macrophages in vitro than their Cd47-negative counterparts. For the generation of human-mouse chimeric livers in immunodeficient BALB-ΔRAG/γ(c) -uPA (urokinase-type plasminogen activator) mice, freshly thawed cryopreserved human hepatocytes were transduced with a lentiviral expression vector for Cd47 using a refined in vitro transduction protocol immediately before transplantation. In vivo, Cd47-positive human primary hepatocytes were selectively retained following engraftment in immunodeficient mice, leading to at least a doubling of liver repopulation efficiencies. CONCLUSION We conclude that ectopic expression of murine Cd47 in human hepatocytes selectively favors engraftment upon transplantation into mice, a finding that should have a profound impact on the generation of robust humanized small animal models. Moreover, dominance of ectopically expressed murine Cd47 over endogenous human CD47 should also widen the spectrum of immunodeficient mouse strains suitable for humanization.
Collapse
Affiliation(s)
- Johan M Waern
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Vaughan AM, Mikolajczak SA, Wilson EM, Grompe M, Kaushansky A, Camargo N, Bial J, Ploss A, Kappe SHI. Complete Plasmodium falciparum liver-stage development in liver-chimeric mice. J Clin Invest 2012; 122:3618-28. [PMID: 22996664 DOI: 10.1172/jci62684] [Citation(s) in RCA: 176] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 07/12/2012] [Indexed: 11/17/2022] Open
Abstract
Plasmodium falciparum, which causes the most lethal form of human malaria, replicates in the host liver during the initial stage of infection. However, in vivo malaria liver-stage (LS) studies in humans are virtually impossible, and in vitro models of LS development do not reconstitute relevant parasite growth conditions. To overcome these obstacles, we have adopted a robust mouse model for the study of P. falciparum LS in vivo: the immunocompromised and fumarylacetoacetate hydrolase-deficient mouse (Fah-/-, Rag2-/-, Il2rg-/-, termed the FRG mouse) engrafted with human hepatocytes (FRG huHep). FRG huHep mice supported vigorous, quantifiable P. falciparum LS development that culminated in complete maturation of LS at approximately 7 days after infection, providing a relevant model for LS development in humans. The infections allowed observations of previously unknown expression of proteins in LS, including P. falciparum translocon of exported proteins 150 (PTEX150) and exported protein-2 (EXP-2), components of a known parasite protein export machinery. LS schizonts exhibited exoerythrocytic merozoite formation and merosome release. Furthermore, FRG mice backcrossed to the NOD background and repopulated with huHeps and human red blood cells supported reproducible transition from LS infection to blood-stage infection. Thus, these mice constitute reliable models to study human LS directly in vivo and demonstrate utility for studies of LS-to-blood-stage transition of a human malaria parasite.
Collapse
Affiliation(s)
- Ashley M Vaughan
- Seattle Biomedical Research Institute, Seattle, Washington 98109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|