51
|
Villaudy J, Schotte R, Legrand N, Spits H. Critical assessment of human antibody generation in humanized mouse models. J Immunol Methods 2014; 410:18-27. [PMID: 24952244 DOI: 10.1016/j.jim.2014.06.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/10/2014] [Accepted: 06/10/2014] [Indexed: 12/31/2022]
Abstract
Immunodeficient mice reconstituted with human hematopoietic stem cells provide a small-animal model for the study of development and function of human hematopoietic cells in vivo. However, in the current models, the immune response, and especially the humoral response by the human immune cells is far from optimal. The B cells found in these mice exhibit an immature and abnormal phenotype correlating with a reduced capacity to produce antigen-specific affinity matured antibodies upon infection or immunization. Herein, we review the current state of knowledge of development, function and antibody production of human B cells and discuss the obstacles for the improvement of these models.
Collapse
Affiliation(s)
- Julien Villaudy
- AIMM Therapeutics, Meibergdreef 59, 1105 BA Amsterdam Zuidoost, Netherlands; Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 BA Amsterdam Zuidoost, Netherlands.
| | - Remko Schotte
- AIMM Therapeutics, Meibergdreef 59, 1105 BA Amsterdam Zuidoost, Netherlands.
| | - Nicolas Legrand
- AXENIS, Institut Pasteur, Centre Francois Jacob, 28, rue du Dr. Roux, 75015 Paris, France.
| | - Hergen Spits
- AIMM Therapeutics, Meibergdreef 59, 1105 BA Amsterdam Zuidoost, Netherlands; Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 BA Amsterdam Zuidoost, Netherlands.
| |
Collapse
|
52
|
Werner-Klein M, Proske J, Werno C, Schneider K, Hofmann HS, Rack B, Buchholz S, Ganzer R, Blana A, Seelbach-Göbel B, Nitsche U, Männel DN, Klein CA. Immune humanization of immunodeficient mice using diagnostic bone marrow aspirates from carcinoma patients. PLoS One 2014; 9:e97860. [PMID: 24830425 PMCID: PMC4022674 DOI: 10.1371/journal.pone.0097860] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/24/2014] [Indexed: 01/18/2023] Open
Abstract
Tumor xenografts in immunodeficient mice, while routinely used in cancer research, preclude studying interactions of immune and cancer cells or, if humanized by allogeneic immune cells, are of limited use for tumor-immunological questions. Here, we explore a novel way to generate cancer models with an autologous humanized immune system. We demonstrate that hematopoietic stem and progenitor cells (HSPCs) from bone marrow aspirates of non-metastasized carcinoma patients, which are taken at specialized centers for diagnostic purposes, can be used to generate a human immune system in NOD-scid IL2rγ(null) (NSG) and HLA-I expressing NSG mice (NSG-HLA-A2/HHD) comprising both, lymphoid and myeloid cell lineages. Using NSG-HLA-A2/HHD mice, we show that responsive and self-tolerant human T cells develop and human antigen presenting cells can activate human T cells. As critical factors we identified the low potential of bone marrow HSPCs to engraft, generally low HSPC numbers in patient-derived bone marrow samples, cryopreservation and routes of cell administration. We provide here an optimized protocol that uses a minimum number of HSPCs, preselects high-quality bone marrow samples defined by the number of initially isolated leukocytes and intra-femoral or intra-venous injection. In conclusion, the use of diagnostic bone marrow aspirates from non-metastasized carcinoma patients for the immunological humanization of immunodeficient mice is feasible and opens the chance for individualized analyses of anti-tumoral T cell responses.
Collapse
Affiliation(s)
| | - Judith Proske
- Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Christian Werno
- Project Group Personalized Tumor Therapy, Fraunhofer Institute of Toxicology and Experimental Medicine, Regensburg, Germany
| | - Katharina Schneider
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | | | - Brigitte Rack
- Department of Gynecology and Obstetrics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stefan Buchholz
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
| | - Roman Ganzer
- Department of Urology, University of Leipzig, Leipzig, Germany
| | - Andreas Blana
- Department of Urology, Fuerth Hospital, Fuerth, Germany
| | - Birgit Seelbach-Göbel
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Ulrich Nitsche
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Daniela N. Männel
- Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Christoph A. Klein
- Project Group Personalized Tumor Therapy, Fraunhofer Institute of Toxicology and Experimental Medicine, Regensburg, Germany
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| |
Collapse
|
53
|
OKT3 prevents xenogeneic GVHD and allows reliable xenograft initiation from unfractionated human hematopoietic tissues. Blood 2014; 123:e134-44. [PMID: 24778156 DOI: 10.1182/blood-2014-02-556340] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Immunodeficient mice are now readily engrafted with human hematopoietic cells. However, these mice are susceptible to graft-versus-host disease (GVHD) induced by the engraftment and rapid expansion of coinjected human T cells. Therefore, highly purified sample populations must be used, adding significant time, expense, and effort. Here, we have explored in vivo and in vitro methods utilizing anti-T-cell antibodies to circumvent this problem. Intraperitoneal injection of the antibody within 48 hours prevented GVHD. Alternatively, short-term in vitro incubation of cells with antibody immediately before transplant was equally effective. Although in vitro antithymocyte globulin treatment resulted in a dramatic loss of SCID-repopulating cells (SRCs), treatment with OKT3 or UCHT1 abrogated GVHD risk and preserved engraftment potential. Leukemia samples that presented with substantial human T-cell contamination were effectively rescued from GVHD. In addition, OKT3 treatment of unfractionated cord blood resulted in robust engraftment of primary and secondary mice that was indistinguishable from grafts obtained using purified CD34(+) cells. Limiting dilution analysis of unfractionated blood demonstrated a SRC frequency of 1 in 300 to 500 CD34(+) cells, similar to that of purified hematopoietic stem and progenitor cells. This protocol streamlines xenograft studies while significantly reducing the cost and time of the procedure.
Collapse
|
54
|
Rongvaux A, Willinger T, Martinek J, Strowig T, Gearty SV, Teichmann LL, Saito Y, Marches F, Halene S, Palucka AK, Manz MG, Flavell RA. Development and function of human innate immune cells in a humanized mouse model. Nat Biotechnol 2014; 32:364-72. [PMID: 24633240 PMCID: PMC4017589 DOI: 10.1038/nbt.2858] [Citation(s) in RCA: 602] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 02/24/2014] [Indexed: 12/22/2022]
Abstract
Mice repopulated with human hematopoietic cells are a powerful tool for the study of human hematopoiesis and immune function in vivo. However, existing humanized mouse models are unable to support development of human innate immune cells, including myeloid cells and NK cells. Here we describe a mouse strain, called MI(S)TRG, in which human versions of four genes encoding cytokines important for innate immune cell development are knocked in to their respective mouse loci. The human cytokines support the development and function of monocytes/macrophages and natural killer cells derived from human fetal liver or adult CD34+ progenitor cells injected into the mice. Human macrophages infiltrated a human tumor xenograft in MI(S)TRG mice in a manner resembling that observed in tumors obtained from human patients. This humanized mouse model may be used to model the human immune system in scenarios of health and pathology, and may enable evaluation of therapeutic candidates in an in vivo setting relevant to human physiology.
Collapse
Affiliation(s)
- Anthony Rongvaux
- 1] Department of Immunobiology, Yale University, New Haven, Connecticut, USA. [2]
| | - Tim Willinger
- 1] Department of Immunobiology, Yale University, New Haven, Connecticut, USA. [2]
| | - Jan Martinek
- 1] Baylor Institute for Immunology Research, Dallas, Texas, USA. [2] Biomedical studies program, Baylor University, Waco, Texas, USA
| | - Till Strowig
- 1] Department of Immunobiology, Yale University, New Haven, Connecticut, USA. [2]
| | - Sofia V Gearty
- Department of Immunobiology, Yale University, New Haven, Connecticut, USA
| | - Lino L Teichmann
- 1] Department of Laboratory Medicine, Yale University, New Haven, Connecticut, USA. [2] Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Yasuyuki Saito
- Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University, New Haven, Connecticut, USA
| | | | - Markus G Manz
- Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Richard A Flavell
- 1] Department of Immunobiology, Yale University, New Haven, Connecticut, USA. [2] Howard Hughes Medical Institute, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
55
|
Covassin L, Jangalwe S, Jouvet N, Laning J, Burzenski L, Shultz LD, Brehm MA. Human immune system development and survival of non-obese diabetic (NOD)-scid IL2rγ(null) (NSG) mice engrafted with human thymus and autologous haematopoietic stem cells. Clin Exp Immunol 2014; 174:372-88. [PMID: 23869841 DOI: 10.1111/cei.12180] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2013] [Indexed: 01/01/2023] Open
Abstract
Immunodeficient mice bearing targeted mutations in the IL2rg gene and engrafted with human immune systems are effective tools for the study of human haematopoiesis, immunity, infectious disease and transplantation biology. The most robust human immune model is generated by implantation of human fetal thymic and liver tissues in irradiated recipients followed by intravenous injection of autologous fetal liver haematopoietic stem cells [often referred to as the BLT (bone marrow, liver, thymus) model]. To evaluate the non-obese diabetic (NOD)-scid IL2rγ(null) (NSG)-BLT model, we have assessed various engraftment parameters and how these parameters influence the longevity of NSG-BLT mice. We observed that irradiation and subrenal capsule implantation of thymus/liver fragments was optimal for generating human immune systems. However, after 4 months, a high number of NSG-BLT mice develop a fatal graft-versus-host disease (GVHD)-like syndrome, which correlates with the activation of human T cells and increased levels of human immunoglobulin (Ig). Onset of GVHD was not delayed in NSG mice lacking murine major histocompatibility complex (MHC) classes I or II and was not associated with a loss of human regulatory T cells or absence of intrathymic cells of mouse origin (mouse CD45(+) ). Our findings demonstrate that NSG-BLT mice develop robust human immune systems, but that the experimental window for these mice may be limited by the development of GVHD-like pathological changes.
Collapse
Affiliation(s)
- L Covassin
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | | | | | | | | | | | | |
Collapse
|
56
|
Abstract
Humanized mice historically have not been good models of human humoral immunity induced by either infection or immunization. However, newer versions of humanized mice generated in severely immunodeficient mice with a targeted disruption of the IL2Rγc gene have recently been reported to produce antigen-specific class-switched human antibodies, with some demonstrating neutralizing activities. Here we review the growing ability of humanized mice to support the study of human humoral immune responses, discussing the current and future potential of these models as well as their current limitations.
Collapse
Affiliation(s)
- Edward Seung
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology
| | | |
Collapse
|
57
|
Bennett MS, Akkina R. Gene therapy strategies for HIV/AIDS: preclinical modeling in humanized mice. Viruses 2013; 5:3119-41. [PMID: 24351796 PMCID: PMC3967164 DOI: 10.3390/v5123119] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/04/2013] [Accepted: 12/03/2013] [Indexed: 12/28/2022] Open
Abstract
In the absence of an effective vaccine and lack of a complete cure, gene therapy approaches to control HIV infection offer feasible alternatives. Due to the chronic nature of infection, a wide window of opportunity exists to gene modify the HIV susceptible cells that continuously arise from the bone marrow source. To evaluate promising gene therapy approaches that employ various anti-HIV therapeutic molecules, an ideal animal model is necessary to generate important efficacy and preclinical data. In this regard, the humanized mouse models that harbor human hematopoietic cells susceptible to HIV infection provide a suitable in vivo system. This review summarizes the currently used humanized mouse models and different anti-HIV molecules utilized for conferring HIV resistance. Humanized mouse models are compared for their utility in this context and provide perspectives for new directions.
Collapse
Affiliation(s)
| | - Ramesh Akkina
- Department of Microbiology, Immunology and Pathology, Colorado State University, 1619 Campus delivery, Fort Collins, CO 80523, USA.
| |
Collapse
|
58
|
Jawa V, Cousens LP, Awwad M, Wakshull E, Kropshofer H, De Groot AS. T-cell dependent immunogenicity of protein therapeutics: Preclinical assessment and mitigation. Clin Immunol 2013; 149:534-55. [PMID: 24263283 DOI: 10.1016/j.clim.2013.09.006] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/13/2013] [Accepted: 09/14/2013] [Indexed: 02/07/2023]
Abstract
Protein therapeutics hold a prominent and rapidly expanding place among medicinal products. Purified blood products, recombinant cytokines, growth factors, enzyme replacement factors, monoclonal antibodies, fusion proteins, and chimeric fusion proteins are all examples of therapeutic proteins that have been developed in the past few decades and approved for use in the treatment of human disease. Despite early belief that the fully human nature of these proteins would represent a significant advantage, adverse effects associated with immune responses to some biologic therapies have become a topic of some concern. As a result, drug developers are devising strategies to assess immune responses to protein therapeutics during both the preclinical and the clinical phases of development. While there are many factors that contribute to protein immunogenicity, T cell- (thymus-) dependent (Td) responses appear to play a critical role in the development of antibody responses to biologic therapeutics. A range of methodologies to predict and measure Td immune responses to protein drugs has been developed. This review will focus on the Td contribution to immunogenicity, summarizing current approaches for the prediction and measurement of T cell-dependent immune responses to protein biologics, discussing the advantages and limitations of these technologies, and suggesting a practical approach for assessing and mitigating Td immunogenicity.
Collapse
|
59
|
Billerbeck E, de Jong Y, Dorner M, de la Fuente C, Ploss A. Animal models for hepatitis C. Curr Top Microbiol Immunol 2013; 369:49-86. [PMID: 23463197 DOI: 10.1007/978-3-642-27340-7_3] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatitis C remains a global epidemic. Approximately 3 % of the world's population suffers from chronic hepatitis C, which is caused by hepatitis C virus (HCV)-a positive sense, single-stranded RNA virus of the Flaviviridae family. HCV has a high propensity for establishing a chronic infection. If untreated chronic HCV carriers can develop severe liver disease including fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Antiviral treatment is only partially effective, costly, and poorly tolerated. A prophylactic or therapeutic vaccine for HCV does not exist. Mechanistic studies of virus-host interactions, HCV immunity, and pathogenesis as well as the development of more effective therapies have been hampered by the lack of a suitable small animal model. Besides humans, chimpanzees are the only species that is naturally susceptible to HCV infection. While experimentation in these large primates has yielded valuable insights, ethical considerations, limited availability, genetic heterogeneity, and cost limit their utility. In search for more tractable small animal models, numerous experimental approaches have been taken to recapitulate parts of the viral life cycle and/or aspects of viral pathogenesis that will be discussed in this review. Exciting new models and improvements in established models hold promise to further elucidate our understanding of chronic HCV infection.
Collapse
Affiliation(s)
- Eva Billerbeck
- Center for the Study of Hepatitis C, The Rockefeller University, NY, USA
| | | | | | | | | |
Collapse
|
60
|
Mailly L, Robinet E, Meuleman P, Baumert TF, Zeisel MB. Hepatitis C virus infection and related liver disease: the quest for the best animal model. Front Microbiol 2013; 4:213. [PMID: 23898329 PMCID: PMC3724122 DOI: 10.3389/fmicb.2013.00212] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 07/08/2013] [Indexed: 12/13/2022] Open
Abstract
Hepatitis C virus (HCV) is a major cause of cirrhosis and hepatocellular carcinoma (HCC) making the virus the most common cause of liver failure and transplantation. HCV is estimated to chronically affect 130 million individuals and to lead to more than 350,000 deaths per year worldwide. A vaccine is currently not available. The recently developed direct acting antivirals (DAAs) have markedly increased the efficacy of the standard of care but are not efficient enough to completely cure all chronically infected patients and their toxicity limits their use in patients with advanced liver disease, co-morbidity or transplant recipients. Because of the host restriction, which is limited to humans and non-human primates, in vivo study of HCV infection has been hampered since its discovery more than 20 years ago. The chimpanzee remains the most physiological model to study the innate and adaptive immune responses, but its use is ethically difficult and is now very restricted and regulated. The development of a small animal model that allows robust HCV infection has been achieved using chimeric liver immunodeficient mice, which are therefore not suitable for studying the adaptive immune responses. Nevertheless, these models allowed to go deeply in the comprehension of virus-host interactions and to assess different therapeutic approaches. The immunocompetent mouse models that were recently established by genetic humanization have shown an interesting improvement concerning the study of the immune responses but are still limited by the absence of the complete robust life cycle of the virus. In this review, we will focus on the relevant available animal models of HCV infection and their usefulness for deciphering the HCV life cycle and virus-induced liver disease, as well as for the development and evaluation of new therapeutics. We will also discuss the perspectives on future immunocompetent mouse models and the hurdles to their development.
Collapse
Affiliation(s)
- Laurent Mailly
- Inserm U1110, Université de Strasbourg Strasbourg, France
| | | | | | | | | |
Collapse
|
61
|
Billerbeck E, Horwitz JA, Labitt RN, Donovan BM, Vega K, Budell WC, Koo GC, Rice CM, Ploss A. Characterization of human antiviral adaptive immune responses during hepatotropic virus infection in HLA-transgenic human immune system mice. THE JOURNAL OF IMMUNOLOGY 2013; 191:1753-64. [PMID: 23833235 DOI: 10.4049/jimmunol.1201518] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Humanized mice have emerged as a promising model to study human immunity in vivo. Although they are susceptible to many pathogens exhibiting an almost exclusive human tropism, human immune responses to infection remain functionally impaired. It has recently been demonstrated that the expression of HLA molecules improves human immunity to lymphotropic virus infections in humanized mice. However, little is known about the extent of functional human immune responses in nonlymphoid tissues, such as in the liver, and the role of HLA expression in this context. Therefore, we analyzed human antiviral immunity in humanized mice during a hepatotropic adenovirus infection. We compared immune responses of conventional humanized NOD SCID IL-2Rγ-deficient (NSG) mice to those of a novel NOD SCID IL-2Rγ-deficient strain transgenic for both HLA-A*0201 and a chimeric HLA-DR*0101 molecule. Using a firefly luciferase-expressing adenovirus and in vivo bioluminescence imaging, we demonstrate a human T cell-dependent partial clearance of adenovirus-infected cells from the liver of HLA-transgenic humanized mice. This correlated with liver infiltration and activation of T cells, as well as the detection of Ag-specific humoral and cellular immune responses. When infected with a hepatitis C virus NS3-expressing adenovirus, HLA-transgenic humanized mice mounted an HLA-A*0201-restricted hepatitis C virus NS3-specific CD8(+) T cell response. In conclusion, our study provides evidence for the generation of partial functional antiviral immune responses against a hepatotropic pathogen in humanized HLA-transgenic mice. The adenovirus reporter system used in our study may serve as simple in vivo method to evaluate future strategies for improving human intrahepatic immune responses in humanized mice.
Collapse
Affiliation(s)
- Eva Billerbeck
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Akkina R. Human immune responses and potential for vaccine assessment in humanized mice. Curr Opin Immunol 2013; 25:403-9. [PMID: 23628166 DOI: 10.1016/j.coi.2013.03.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 03/27/2013] [Indexed: 12/13/2022]
Abstract
The new humanized mouse models with a transplanted human immune system have a capacity for de novo multilineage human hematopoiesis and generate T cells, B cells, macrophages, dendritic cells and NK cells. Of the two current leading humanized mouse models, the hu-HSC model is created by human hematopoietic stem cell (HSC) engraftment whereas the BLT mouse model is prepared by co-transplantation of human fetal liver, thymus and HSC. Humoral and cellular immune responses are seen in both models after immunization with antigens or infection with hematotropic pathogens such as EBV, HIV-1 and dengue viruses. While consistent antigen specific IgM production is seen, IgG responses were found to be generally feeble which is attributed to inefficient immunoglobulin class switching. BLT mice permit human HLA restricted T cell responses due to the autologous human thymus contributing to T cell maturation. Use of HLA Class I and II transgenic hu-HSC mice recently demonstrated that the HLA restriction deficiency could be overcome in this model. However, the overall vigor of the immune responses needs further improvement in both the models to approach that of the human. Towards this goal, supplementation with human cytokines and growth factors by transgenesis to improve human cell reconstitution and their homeostatic maintenance are beginning to yield improved mouse strains to create more robust human immune competent mice for immunoprophylaxis studies.
Collapse
Affiliation(s)
- Ramesh Akkina
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
63
|
Fujiwara S, Matsuda G, Imadome KI. Humanized mouse models of epstein-barr virus infection and associated diseases. Pathogens 2013; 2:153-76. [PMID: 25436886 PMCID: PMC4235711 DOI: 10.3390/pathogens2010153] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 02/26/2013] [Accepted: 03/05/2013] [Indexed: 12/29/2022] Open
Abstract
Epstein-Barr virus (EBV) is a ubiquitous herpesvirus infecting more than 90% of the adult population of the world. EBV is associated with a variety of diseases including infectious mononucleosis, lymphoproliferative diseases, malignancies such as Burkitt lymphoma and nasopharyngeal carcinoma, and autoimmune diseases including rheumatoid arthritis (RA). EBV in nature infects only humans, but in an experimental setting, a limited species of new-world monkeys can be infected with the virus. Small animal models, suitable for evaluation of novel therapeutics and vaccines, have not been available. Humanized mice, defined here as mice harboring functioning human immune system components, are easily infected with EBV that targets cells of the hematoimmune system. Furthermore, humanized mice can mount both cellular and humoral immune responses to EBV. Thus, many aspects of human EBV infection, including associated diseases (e.g., lymphoproliferative disease, hemophagocytic lymphohistiocytosis and erosive arthritis resembling RA), latent infection, and T-cell-mediated and humoral immune responses have been successfully reproduced in humanized mice. Here we summarize recent achievements in the field of humanized mouse models of EBV infection and show how they have been utilized to analyze EBV pathogenesis and normal and aberrant human immune responses to the virus.
Collapse
Affiliation(s)
- Shigeyoshi Fujiwara
- Department of Infectious Diseases, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan.
| | - Go Matsuda
- Department of Infectious Diseases, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan.
| | - Ken-Ichi Imadome
- Department of Infectious Diseases, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan.
| |
Collapse
|
64
|
Tiniakou E, Costenbader KH, Kriegel MA. Sex-specific environmental influences on the development of autoimmune diseases. Clin Immunol 2013; 149:182-91. [PMID: 23507400 DOI: 10.1016/j.clim.2013.02.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 02/12/2013] [Accepted: 02/13/2013] [Indexed: 10/27/2022]
Abstract
Sex differences in autoimmune diseases are evolutionarily tied to the fact that the female immune system is confronted with intense alterations during menstrual cycles, pregnancy and childbirth. These events may be associated with breaches in the mucosal epithelial layers that are shielding us from environmental factors. Associations between environmental agents and autoimmune diseases have been described extensively in prior studies. Little evidence, however, exists for sex-specific environmental effects on autoimmune diseases. In this review, we summarize studies involving this often-neglected aspect. We give examples of environmental factors that may influence the sex bias in autoimmunity. We conclude that most studies do not give insight into sex-specific environmental effects due to the influence of gender-selective social, occupational or other exposures. Prospective studies are needed in order to determine true sex-biased environmental influences. Finally, humanized murine models might aid in better understanding the mechanisms involved in sex-specific environmental effects on autoimmune diseases.
Collapse
Affiliation(s)
- Eleni Tiniakou
- Department of Immunobiology, Yale University School of Medicine, CT 06510, USA
| | | | | |
Collapse
|
65
|
Increasing hematopoietic stem cell yield to develop mice with human immune systems. BIOMED RESEARCH INTERNATIONAL 2013; 2013:740892. [PMID: 23509770 PMCID: PMC3586441 DOI: 10.1155/2013/740892] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 12/17/2012] [Accepted: 12/27/2012] [Indexed: 01/14/2023]
Abstract
Hematopoietic stem cells (HSCs) are unique in their capacity to give rise to all mature cells of the immune system. For years, HSC transplantation has been used for treatment of genetic and neoplastic diseases of the hematopoietic and immune systems. The sourcing of HSCs from human umbilical cord blood has salient advantages over isolation from mobilized peripheral blood. However, poor sample yield has prompted development of methodologies to expand HSCs ex vivo. Cytokines, trophic factors, and small molecules have been variously used to promote survival and proliferation of HSCs in culture, whilst strategies to lower the concentration of inhibitors in the culture media have recently been applied to promote HSC expansion. In this paper, we outline strategies to expand HSCs in vitro, and to improve engraftment and reconstitution of human immune systems in immunocompromised mice. To the extent that these “humanized” mice are representative of the endogenous human immune system, they will be invaluable tools for both basic science and translational medicine.
Collapse
|
66
|
Engineering of a functional bone organ through endochondral ossification. Proc Natl Acad Sci U S A 2013; 110:3997-4002. [PMID: 23401508 DOI: 10.1073/pnas.1220108110] [Citation(s) in RCA: 244] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Embryonic development, lengthening, and repair of most bones proceed by endochondral ossification, namely through formation of a cartilage intermediate. It was previously demonstrated that adult human bone marrow-derived mesenchymal stem/stromal cells (hMSCs) can execute an endochondral program and ectopically generate mature bone. Here we hypothesized that hMSCs pushed through endochondral ossification can engineer a scaled-up ossicle with features of a "bone organ," including physiologically remodeled bone, mature vasculature, and a fully functional hematopoietic compartment. Engineered hypertrophic cartilage required IL-1β to be efficiently remodeled into bone and bone marrow upon subcutaneous implantation. This model allowed distinguishing, by analogy with bone development and repair, an outer, cortical-like perichondral bone, generated mainly by host cells and laid over a premineralized area, and an inner, trabecular-like, endochondral bone, generated mainly by the human cells and formed over the cartilaginous template. Hypertrophic cartilage remodeling was paralleled by ingrowth of blood vessels, displaying sinusoid-like structures and stabilized by pericytic cells. Marrow cavities of the ossicles contained phenotypically defined hematopoietic stem cells and progenitor cells at similar frequencies as native bones, and marrow from ossicles reconstituted multilineage long-term hematopoiesis in lethally irradiated mice. This study, by invoking a "developmental engineering" paradigm, reports the generation by appropriately instructed hMSC of an ectopic "bone organ" with a size, structure, and functionality comparable to native bones. The work thus provides a model useful for fundamental and translational studies of bone morphogenesis and regeneration, as well as for the controlled manipulation of hematopoietic stem cell niches in physiology and pathology.
Collapse
|
67
|
Akkina R. New generation humanized mice for virus research: comparative aspects and future prospects. Virology 2013; 435:14-28. [PMID: 23217612 DOI: 10.1016/j.virol.2012.10.007] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 10/03/2012] [Accepted: 10/03/2012] [Indexed: 12/27/2022]
Abstract
Work with human specific viruses will greatly benefit from the use of an in vivo system that provides human target cells and tissues in a physiological setting. In this regard humanized mice (hu-Mice) have played an important role in our understanding of viral pathogenesis and testing of therapeutic strategies. Limitations with earlier versions of hu-Mice that lacked a functioning human immune system are currently being overcome. The new generation hu-Mouse models are capable of multilineage human hematopoiesis and generate T cells, B cells, macrophages and dendritic cells required for an adaptive human immune response. Now any human specific pathogen that can infect humanized mice can be studied in the context of ongoing infection and immune responses. Two leading humanized mouse models are currently employed: the hu-HSC model is created by transplantation of human hematopoietic stem cells (HSC), whereas the BLT mouse model is prepared by transplantation of human fetal liver, thymus and HSC. A number of human specific viruses such as HIV-1, dengue, EBV and HCV are being studied intensively in these systems. Both models permit infection by mucosal routes with viruses such as HIV-1 thus allowing transmission prevention studies. Cellular and humoral immune responses are seen in both the models. While there is efficient antigen specific IgM production, IgG responses are suboptimal due to inefficient immunoglobulin class switching. With the maturation of T cells occurring in the autologous human thymus, BLT mice permit human HLA restricted T cell responses in contrast to hu-HSC mice. However, the strength of the immune responses needs further improvement in both models to reach the levels seen in humans. The scope of hu-Mice use is further broadened by transplantation of additional tissues like human liver thus permitting immunopathogenesis studies on hepatotropic viruses such as HCV. Numerous studies that encompass antivirals, gene therapy, viral evolution, and the generation of human monoclonal antibodies have been conducted with promising results in these mice. For further improvement of the new hu-Mouse models, ongoing work is focused on generating new strains of immunodeficient mice transgenic for human HLA molecules to strengthen immune responses and human cytokines and growth factors to improve human cell reconstitution and their homeostatic maintenance.
Collapse
Affiliation(s)
- Ramesh Akkina
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
68
|
Sandmann L, Ploss A. Barriers of hepatitis C virus interspecies transmission. Virology 2013; 435:70-80. [PMID: 23217617 PMCID: PMC3523278 DOI: 10.1016/j.virol.2012.09.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 09/28/2012] [Indexed: 12/19/2022]
Abstract
Hepatitis C virus (HCV) is a major causative agent of severe liver disease including fibrosis, cirrhosis and liver cancer. Therapy has improved over the years, but continues to be associated with adverse side effects and variable success rates. Furthermore, a vaccine protecting against HCV infection remains elusive. Development of more effective intervention measures has been delayed by the lack of a suitable animal model. Naturally, HCV infects only humans and chimpanzees. The determinants of this limited host range are poorly understood in part due to difficulties of studying HCV in cell culture. Some progress has been made elucidating the barriers for the HCV lifecycle in non-permissive species which will help in the future to construct animal models for HCV infection, immunity and pathogenesis.
Collapse
|
69
|
Lang J, Kelly M, Freed BM, McCarter MD, Kedl RM, Torres RM, Pelanda R. Studies of lymphocyte reconstitution in a humanized mouse model reveal a requirement of T cells for human B cell maturation. THE JOURNAL OF IMMUNOLOGY 2013; 190:2090-101. [PMID: 23335750 DOI: 10.4049/jimmunol.1202810] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The hematopoietic humanized mouse (hu-mouse) model is a powerful resource to study and manipulate the human immune system. However, a major and recurrent issue with this model has been the poor maturation of B cells that fail to progress beyond the transitional B cell stage. Of interest, a similar problem has been reported in transplant patients who receive cord blood stem cells. In this study, we characterize the development of human B and T cells in the lymph nodes (LNs) and spleen of BALB/c-Rag2(null)Il2rγ(null) hu-mice. We find a dominant population of immature B cells in the blood and spleen early, followed by a population of human T cells, coincident with the detection of LNs. Notably, in older mice we observe a major population of mature B cells in LNs and in the spleens of mice with higher T cell frequencies. Moreover, we demonstrate that T cells are necessary for B cell maturation, as introduction of autologous human T cells expedites the appearance of mature B cells, whereas in vivo depletion of T cells retards B cell maturation. The presence of the mature B cell population correlates with enhanced IgG and Ag-specific responses to both T cell-dependent and T cell-independent challenges, indicating their functionality. These findings enhance our understanding of human B cell development, provide increased details of the reconstitution dynamics of hu-mice, and validate the use of this animal model to study mechanisms and treatments for the similar delay of functional B cells associated with cord blood transplantations.
Collapse
Affiliation(s)
- Julie Lang
- Integrated Department of Immunology, National Jewish Health, Denver, CO 80206, USA
| | | | | | | | | | | | | |
Collapse
|
70
|
Rongvaux A, Takizawa H, Strowig T, Willinger T, Eynon EE, Flavell RA, Manz MG. Human hemato-lymphoid system mice: current use and future potential for medicine. Annu Rev Immunol 2013; 31:635-674. [PMID: 23330956 DOI: 10.1146/annurev-immunol-032712-095921] [Citation(s) in RCA: 267] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To directly study complex human hemato-lymphoid system physiology and respective system-associated diseases in vivo, human-to-mouse xenotransplantation models for human blood and blood-forming cells and organs have been developed over the past three decades. We here review the fundamental requirements and the remarkable progress made over the past few years in improving these systems, the current major achievements reached by use of these models, and the future challenges to more closely model and study human health and disease and to achieve predictive preclinical testing of both prevention measures and potential new therapies.
Collapse
Affiliation(s)
- Anthony Rongvaux
- Department of Immunobiology, Yale University, New Haven, Connecticut 06520
| | - Hitoshi Takizawa
- Division of Hematology, University Hospital Zürich, CH-8091 Zürich, Switzerland
| | - Till Strowig
- Department of Immunobiology, Yale University, New Haven, Connecticut 06520
| | - Tim Willinger
- Department of Immunobiology, Yale University, New Haven, Connecticut 06520
| | - Elizabeth E Eynon
- Department of Immunobiology, Yale University, New Haven, Connecticut 06520
| | - Richard A Flavell
- Department of Immunobiology, Yale University, New Haven, Connecticut 06520.,Howard Hughes Medical Institute, Yale University, New Haven, Connecticut 06520;
| | - Markus G Manz
- Division of Hematology, University Hospital Zürich, CH-8091 Zürich, Switzerland
| |
Collapse
|
71
|
Csaszar E, Cohen S, Zandstra PW. Blood stem cell products: Toward sustainable benchmarks for clinical translation. Bioessays 2013; 35:201-10. [DOI: 10.1002/bies.201200118] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
72
|
Hematopoietic Stem Cells. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
73
|
Konantz M, Balci TB, Hartwig UF, Dellaire G, André MC, Berman JN, Lengerke C. Zebrafish xenografts as a tool for in vivo studies on human cancer. Ann N Y Acad Sci 2012; 1266:124-37. [PMID: 22901264 DOI: 10.1111/j.1749-6632.2012.06575.x] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The zebrafish has become a powerful vertebrate model for genetic studies of embryonic development and organogenesis and increasingly for studies in cancer biology. Zebrafish facilitate the performance of reverse and forward genetic approaches, including mutagenesis and small molecule screens. Moreover, several studies report the feasibility of xenotransplanting human cells into zebrafish embryos and adult fish. This model provides a unique opportunity to monitor tumor-induced angiogenesis, invasiveness, and response to a range of treatments in vivo and in real time. Despite the high conservation of gene function between fish and humans, concern remains that potential differences in zebrafish tissue niches and/or missing microenvironmental cues could limit the relevance and translational utility of data obtained from zebrafish human cancer cell xenograft models. Here, we summarize current data on xenotransplantation of human cells into zebrafish, highlighting the advantages and limitations of this model in comparison to classical murine models of xenotransplantation.
Collapse
Affiliation(s)
- Martina Konantz
- Department of Hematology and Oncology, University of Tübingen Medical Center II, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
74
|
Shultz LD, Brehm MA, Garcia-Martinez JV, Greiner DL. Humanized mice for immune system investigation: progress, promise and challenges. Nat Rev Immunol 2012; 12:786-98. [PMID: 23059428 DOI: 10.1038/nri3311] [Citation(s) in RCA: 725] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significant advances in our understanding of the in vivo functions of human cells and tissues and the human immune system have resulted from the development of 'humanized' mouse strains that are based on severely immunodeficient mice with mutations in the interleukin-2 receptor common γ-chain locus. These mouse strains support the engraftment of a functional human immune system and permit detailed analyses of human immune biology, development and functions. In this Review, we discuss recent advances in the development and utilization of humanized mice, the lessons learnt, the remaining challenges and the promise of using humanized mice for the in vivo study of human immunology.
Collapse
Affiliation(s)
- Leonard D Shultz
- Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA.
| | | | | | | |
Collapse
|
75
|
Dodon MD, Villaudy J, Gazzolo L, Haines R, Lairmore M. What we are learning on HTLV-1 pathogenesis from animal models. Front Microbiol 2012; 3:320. [PMID: 22969759 PMCID: PMC3431546 DOI: 10.3389/fmicb.2012.00320] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/16/2012] [Indexed: 11/24/2022] Open
Abstract
Isolated and identified more than 30 years ago, human T cell leukemia virus type 1 (HTLV-1) is the etiological agent of adult T cell leukemia/lymphoma, an aggressive lymphoproliferative disease of activated CD4+ T cells, and other inflammatory disorders such as HTLV-1-associated myelopathy/tropical spastic paraparesis. A variety of animal models have contributed to the fundamental knowledge of HTLV-1 transmission, pathogenesis, and to the design of novel therapies to treat HTLV-1-associated diseases. Small animal models (rabbits, rats, and mice) as well as large animal models (monkeys) have been utilized to significantly advance characterization of the viral proteins and of virus-infected cells in the early steps of infection, as well as in the development of leukemogenic and immunopathogenic processes. Over the past two decades, the creation of new immunocompromised mouse strains that are robustly reconstituted with a functional human immune system (HIS) after being transplanted with human tissues or progenitor cells has revolutionized the in vivo investigation of viral infection and pathogenesis. Recent observations obtained in HTLV-1-infected humanized HIS mice that develop lymphomas provide the opportunity to study the evolution of the proviral clonality in human T cells present in different lymphoid organs. Current progress in the improvement of those humanized models will favor the testing of drugs and the development of targeted therapies against HTLV-1-associated diseases.
Collapse
Affiliation(s)
- Madeleine Duc Dodon
- Laboratoire de Biologie Moléculaire de la Cellule, Unité Mixte de Recherche 5239, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon Lyon, France
| | | | | | | | | |
Collapse
|
76
|
Generation of a humanized mouse model with both human immune system and liver cells to model hepatitis C virus infection and liver immunopathogenesis. Nat Protoc 2012; 7:1608-17. [PMID: 22899330 DOI: 10.1038/nprot.2012.083] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Establishing a small animal model that accurately recapitulates hepatotropic pathogens, including hepatitis C virus (HCV) infection and immunopathogenesis, is essential for the study of hepatitis virus-induced liver disease and for therapeutics development. This protocol describes our recently developed humanized mouse model for studying HCV and other hepatotropic infections, human immune response and hepatitis and liver fibrosis. The first 5-h stage is the isolation of human liver progenitor and hematopoietic stem cells from fetal liver. Next, AFC8 immunodeficient mice are transplanted with the isolated progenitor/stem cells. This generally takes 2 h. The transplanted mice are then treated for a month with the mouse liver apoptosis-inducing AFC8 dimerizer and left for an additional 2-month period to permit human liver and immune cell growth as well as system reconstitution and development before inoculation with HCV clinical isolates. HCV infection, human immune response and liver disease are observed with high incidence from approximately 2 months after inoculation.
Collapse
|
77
|
A germline-competent embryonic stem cell line from NOD.Cg-Prkdc scid Il2rg tm1Wjl /SzJ (NSG) mice. Transgenic Res 2012; 22:179-85. [DOI: 10.1007/s11248-012-9629-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 06/18/2012] [Indexed: 01/22/2023]
|
78
|
Abstract
Despite its complexity, blood is probably the best understood developmental system, largely due to seminal experimentation in the mouse. Clinically, hematopoietic stem cell (HSC) transplantation represents the most widely deployed regenerative therapy, but human HSCs have only been characterized relatively recently. The discovery that immune-deficient mice could be engrafted with human cells provided a powerful approach for studying HSCs. We highlight 2 decades of studies focusing on isolation and molecular regulation of human HSCs, therapeutic applications, and early lineage commitment steps, and compare mouse and humanized models to identify both conserved and species-specific mechanisms that will aid future preclinical research.
Collapse
Affiliation(s)
- Sergei Doulatov
- Division of Stem Cell and Developmental Biology, Campbell Family Institute for Cancer Research/Ontario Cancer Institute, Toronto, ON M5G 1L7, Canada
| | | | | | | |
Collapse
|
79
|
Gorantla S, Gendelman HE, Poluektova LY. Can humanized mice reflect the complex pathobiology of HIV-associated neurocognitive disorders? J Neuroimmune Pharmacol 2012; 7:352-62. [PMID: 22222956 PMCID: PMC3782112 DOI: 10.1007/s11481-011-9335-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 12/14/2011] [Indexed: 01/22/2023]
Abstract
There is a rebirth of humanized mouse models in reflecting human immunodeficiency virus (HIV) pathobiology. This has allowed new investigations of viral diversity, immunity and developmental therapeutics. In the past, HIV infection and disease were, in part, mirrored in immune deficient mice reconstituted with human hematopoietic stem cells. What remained from early studies reflected the ability to mirror central nervous system (CNS) disease. As the wide spread use of combination antiretroviral therapies has changed the severity, but not prevalence, of HIV-associated neurocognitive disorders (HAND), mimicking such virus-induced CNS morbidities in humanized animals is essential for HIV/AIDS research activities. To this end, we now review the evidence for how and under what circumstances humanized mice may be utilized for studies of HIV-1 neuropathogenesis.
Collapse
Affiliation(s)
- Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience and Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | |
Collapse
|
80
|
Legrand N, van der Velden GJ, Fang RHT, Douaisi M, Weijer K, Das AT, Blom B, Uittenbogaart CH, Berkhout B, Centlivre M. A doxycycline-dependent human immunodeficiency virus type 1 replicates in vivo without inducing CD4+ T-cell depletion. J Gen Virol 2012; 93:2017-2027. [PMID: 22647372 DOI: 10.1099/vir.0.042796-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A novel genetic approach for the control of virus replication was used for the design of a conditionally replicating human immunodeficiency virus (HIV) variant, HIV-rtTA. HIV-rtTA gene expression and virus replication are strictly dependent on the presence of a non-toxic effector molecule, doxycycline (dox), and thus can be turned on and off at will in a graded and reversible manner. The in vivo replication capacity, pathogenicity and genetic stability of this HIV-rtTA variant were evaluated in a humanized mouse model of haematopoiesis that harbours lymphoid and myeloid components of the human immune system (HIS). Infection of dox-fed BALB Rag/γc HIS (BRG-HIS) mice with HIV-rtTA led to the establishment of a productive infection without CD4(+) T-cell depletion. The virus did not show any sign of escape from dox control for up to 10 weeks after the onset of infection. No reversion towards a functional Tat-transactivating responsive (TAR) RNA element axis was observed, confirming the genetic stability of the HIV-rtTA variant in vivo. These results demonstrate the proof of concept that HIV-rtTA replicates efficiently in vivo. HIV-rtTA is a promising tool for fundamental research to study virus-host interactions in vivo in a controlled fashion.
Collapse
Affiliation(s)
- Nicolas Legrand
- Department of Cell Biology and Histology, Center for Immunology of Amsterdam (CIA), Academic Medical Center of the University of Amsterdam (AMC-UvA), Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Gisela J van der Velden
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam (AMC-UvA), Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Raphaël Ho Tsong Fang
- Microbiology, Immunology and Molecular Genetics, and Pediatrics, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, USA
| | - Marc Douaisi
- Microbiology, Immunology and Molecular Genetics, and Pediatrics, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, USA
| | - Kees Weijer
- Department of Cell Biology and Histology, Center for Immunology of Amsterdam (CIA), Academic Medical Center of the University of Amsterdam (AMC-UvA), Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Atze T Das
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam (AMC-UvA), Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Bianca Blom
- Department of Cell Biology and Histology, Center for Immunology of Amsterdam (CIA), Academic Medical Center of the University of Amsterdam (AMC-UvA), Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Christel H Uittenbogaart
- Microbiology, Immunology and Molecular Genetics, and Pediatrics, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, USA
| | - Ben Berkhout
- Department of Cell Biology and Histology, Center for Immunology of Amsterdam (CIA), Academic Medical Center of the University of Amsterdam (AMC-UvA), Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Mireille Centlivre
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam (AMC-UvA), Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
81
|
Chang H, Biswas S, Tallarico AS, Sarkis PTN, Geng S, Panditrao MM, Zhu Q, Marasco WA. Human B-cell ontogeny in humanized NOD/SCID γc(null) mice generates a diverse yet auto/poly- and HIV-1-reactive antibody repertoire. Genes Immun 2012; 13:399-410. [PMID: 22592523 PMCID: PMC3411862 DOI: 10.1038/gene.2012.16] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Characterization of the human antibody (Ab) repertoire in mouse models of the human immune system is essential to establish their relevance in translational studies. Single human B-cells were sorted from bone marrow and periphery of humanized NOD/SCID γcnull mice at 8–10 months post-engraftment with human cord blood-derived CD34+ stem cells. Human immunoglobulin variable heavy (VH) and kappa (Vκ) genes were amplified, cognate VH-Vκ gene-pairs assembled as single-chain variable fragment-Fc antibodies (scFvFcs) and functional studies performed. Although overall distribution of VH genes approximated the normal human Ab repertoire, analysis of the VH-third complementarity determining regions (H-CDR3) in the mature B-cell subset demonstrated an increase in length and positive charges suggesting autoimmune characteristics. Additionally, >70% of Vκ sequences utilized Vκ4-1, a germline gene associated with autoimmunity. The mature B-cell subset-derived scFvFcs displayed the highest frequency of autoreactivity and polyspecificity, suggesting defects in checkpoint control mechanisms. Furthermore, these scFvFcs demonstrated binding to recombinant HIV envelope corroborating previous observations of poly/autoreactivity in anti-HIVgp140 antibodies. These data lend support to the hypothesis that anti-HIV BnAbs may be derived from auto/polyspecific Abs that escaped immune elimination and that the hNSG mouse could provide a new experimental platform for studying the origin of anti-HIV neutralizing Ab responses.
Collapse
Affiliation(s)
- H Chang
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Su L. Studying human immunology and immunopathology in humanized mice transplanted with human lymphoid tissues and immune cells. Cell Mol Immunol 2012; 9:191-2. [PMID: 22565218 DOI: 10.1038/cmi.2012.4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
83
|
Ippolito GC, Hoi KH, Reddy ST, Carroll SM, Ge X, Rogosch T, Zemlin M, Shultz LD, Ellington AD, VanDenBerg CL, Georgiou G. Antibody repertoires in humanized NOD-scid-IL2Rγ(null) mice and human B cells reveals human-like diversification and tolerance checkpoints in the mouse. PLoS One 2012; 7:e35497. [PMID: 22558161 PMCID: PMC3338711 DOI: 10.1371/journal.pone.0035497] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/19/2012] [Indexed: 11/25/2022] Open
Abstract
Immunodeficient mice reconstituted with human hematopoietic stem cells enable the in vivo study of human hematopoiesis. In particular, NOD-scid-IL2Rγnull engrafted mice have been shown to have reasonable levels of T and B cell repopulation and can mount T-cell dependent responses; however, antigen-specific B-cell responses in this model are generally poor. We explored whether developmental defects in the immunoglobulin gene repertoire might be partly responsible for the low level of antibody responses in this model. Roche 454 sequencing was used to obtain over 685,000 reads from cDNA encoding immunoglobulin heavy (IGH) and light (IGK and IGL) genes isolated from immature, naïve, or total splenic B cells in engrafted NOD-scid-IL2Rγnull mice, and compared with over 940,000 reads from peripheral B cells of two healthy volunteers. We find that while naïve B-cell repertoires in humanized mice are chiefly indistinguishable from those in human blood B cells, and display highly correlated patterns of immunoglobulin gene segment use, the complementarity-determining region H3 (CDR-H3) repertoires are nevertheless extremely diverse and are specific for each individual. Despite this diversity, preferential DH-JH pairings repeatedly occur within the CDR-H3 interval that are strikingly similar across all repertoires examined, implying a genetic constraint imposed on repertoire generation. Moreover, CDR-H3 length, charged amino-acid content, and hydropathy are indistinguishable between humans and humanized mice, with no evidence of global autoimmune signatures. Importantly, however, a statistically greater usage of the inherently autoreactive IGHV4-34 and IGKV4-1 genes was observed in the newly formed immature B cells relative to naïve B or total splenic B cells in the humanized mice, a finding consistent with the deletion of autoreactive B cells in humans. Overall, our results provide evidence that key features of the primary repertoire are shaped by genetic factors intrinsic to human B cells and are principally unaltered by differences between mouse and human stromal microenvironments.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/genetics
- Antibodies, Monoclonal, Humanized/immunology
- B-Lymphocytes/immunology
- Base Sequence
- Computational Biology
- DNA Primers/genetics
- DNA, Complementary/genetics
- Flow Cytometry
- Fluorescent Dyes
- Genetic Variation
- Hematopoiesis/immunology
- Hematopoietic Stem Cell Transplantation
- Humans
- Immunoglobulin Subunits/genetics
- Interleukin Receptor Common gamma Subunit/genetics
- Mice
- Mice, Inbred NOD/genetics
- Mice, Inbred NOD/immunology
- Mice, SCID/genetics
- Mice, SCID/immunology
- Molecular Sequence Data
- Sequence Analysis, DNA
- Statistics, Nonparametric
Collapse
Affiliation(s)
- Gregory C. Ippolito
- Section of Molecular Genetics and Microbiology, University of Texas at Austin, Austin, Texas, United States of America
| | - Kam Hon Hoi
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
| | - Sai T. Reddy
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Sean M. Carroll
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, United States of America
| | - Xin Ge
- Chemical and Environmental Engineering, University of California, Riverside, California, United States of America
| | - Tobias Rogosch
- Department of Pediatrics, Philips-University, Marburg, Germany
| | - Michael Zemlin
- Department of Pediatrics, Philips-University, Marburg, Germany
| | | | - Andrew D. Ellington
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - Carla L. VanDenBerg
- Department of Pharmacology and Toxicology, University of Texas at Austin, Austin, Texas, United States of America
| | - George Georgiou
- Section of Molecular Genetics and Microbiology, University of Texas at Austin, Austin, Texas, United States of America
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, United States of America
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
84
|
Abstract
In recent years, the technology of constructing chimeric mice with humanized immune systems has markedly improved. Multiple lineages of human immune cells develop in immunodeficient mice that have been transplanted with human hematopoietic stem cells. More importantly, these mice mount functional humoral and cellular immune responses upon immunization or microbial infection. Human immunodeficiency virus type I (HIV-1) can establish an infection in humanized mice, resulting in CD4(+) T-cell depletion and an accompanying nonspecific immune activation, which mimics the immunopathology in HIV-1-infected human patients. This makes humanized mice an optimal model for studying the mechanisms of HIV-1 immunopathogenesis and for developing novel immune-based therapies.
Collapse
|
85
|
Suzuki M, Takahashi T, Katano I, Ito R, Ito M, Harigae H, Ishii N, Sugamura K. Induction of human humoral immune responses in a novel HLA-DR-expressing transgenic NOD/Shi-scid/γcnull mouse. Int Immunol 2012; 24:243-52. [PMID: 22402880 DOI: 10.1093/intimm/dxs045] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mounting evidence has demonstrated that NOD-Shi/scid/γc(null) (NOG) mice are one of the most suitable mouse strains for humanized mouse technologies, in which various human cells or tissues can be engrafted without rejection and autonomously maintained. We have characterized and analyzed various features of the human immune system reconstituted in NOG mice by transplanting human hematopoietic stem cells (hu-HSC). One of the problems of the quasi-immune system in these hu-HSC NOG mice is that the quality of immune responses is not always sufficient, as demonstrated by the lack of IgG production in response to antigen challenge. In this study, we established a novel transgenic NOG sub-strain of mice bearing the HLA-DRA and HLA-DRB1:0405 genes, which specifically expresses HLA-DR4 molecules in MHC II-positive cells. This mouse strain enabled us to match the haplotype of HLA-DR between the recipient mice and human donor HSC. We demonstrated that T-cell homeostasis was differentially regulated in HLA-matched hu-HSC NOG mice compared with HLA-mismatched control mice, and antibody class switching was induced after immunization with exogenous antigens in HLA-matched mice. This novel mouse strain improves the reconstituted human immune systems that develop in humanized mice and will contribute to future studies of human humoral immune responses.
Collapse
Affiliation(s)
- Makiko Suzuki
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Brehm MA, Shultz LD. Human allograft rejection in humanized mice: a historical perspective. Cell Mol Immunol 2012; 9:225-31. [PMID: 22327213 DOI: 10.1038/cmi.2011.64] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Basic research in transplantation immunology has relied primarily on rodent models. Experimentation with rodents has laid the foundation for our basic understanding of the biological events that precipitate rejection of non-self or allogeneic tissue transplants and supported the development of novel strategies to specifically suppress allogeneic immune responses. However, translation of these studies to the clinic has met with limited success, emphasizing the need for new models that focus on human immune responses to allogeneic tissues. Humanized mouse models are an exciting alternative that permits investigation of the rejection of human tissues mediated by human immune cells without putting patients at risk. However, the use of humanized mice is complicated by a diversity of protocols and approaches, including the large number of immunodeficient mouse strains available, the choice of tissue to transplant and the specific human immune cell populations that can be engrafted. Here, we present a historical perspective on the study of allograft rejection in humanized mice and discuss the use of these novel model systems in transplant biology.
Collapse
Affiliation(s)
- Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | | |
Collapse
|
87
|
Abstract
Humanized mouse models that have received human cells or tissue transplants are extremely useful in basic and applied human disease research. Highly immunodeficient mice, which do not reject xenografts and support cell and tissue differentiation and growth, are indispensable for generating additional appropriate models. Since the early 2000s, a series of immunodeficient mice appropriate for generating humanized mice has been successively developed by introducing the IL-2Rγ(null) gene (e.g., NOD/SCID/γc(null) and Rag2(null)γc(null) mice). These strains show not only a high rate of human cell engraftment, but also generate well-differentiated multilineage human hematopoietic cells after human hematopoietic stem cell (HSC) transplantation. These humanized mice facilitate the analysis of human hematology and immunology in vivo. However, human hematopoietic cells developed from HSCs are not always phenotypically and functionally identical to those in humans. More recently, a new series of immunodeficient mice compensates for these disadvantages. These mice were generated by genetically introducing human cytokine genes into NOD/SCID/γc(null) and Rag2(null)γc(null) mice. In this review, we describe the current knowledge of human hematopoietic cells developed in these mice. Various human disease mouse models using these humanized mice are summarized.
Collapse
|
88
|
Human lymphohematopoietic reconstitution and immune function in immunodeficient mice receiving cotransplantation of human thymic tissue and CD34(+) cells. Cell Mol Immunol 2012; 9:232-6. [PMID: 22307039 DOI: 10.1038/cmi.2011.63] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Small animal models with functional human lymphohematopoietic systems are highly valuable for the study of human immune function under physiological and pathological conditions. Over the last two decades, numerous efforts have been devoted towards the development of such humanized mouse models. This review is focused on human lymphohematopoietic reconstitution and immune function in humanized mice by cotransplantation of human fetal thymic tissue and CD34(+) cells. The potential use of these humanized mice in translational biomedical research is also discussed.
Collapse
|
89
|
Cachat A, Villaudy J, Rigal D, Gazzolo L, Duc Dodon M. [Mice are not Men and yet… how humanized mice inform us about human infectious diseases]. Med Sci (Paris) 2012; 28:63-8. [PMID: 22289832 DOI: 10.1051/medsci/2012281018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The study of human pathologies is often limited by the absence of animal models which are robust, cost-effective and reproduce the hallmarks of human infections. While mice have been frequently employed to study human diseases, many of important pathogens display unique human tropism. These last two decades the graft of human progenitor cells or tissues into -immunodeficient mice has allowed the elaboration of so called humanized mice. Humanized mouse technology has made rapid progress, and it is now possible to achieve high levels of human chimerism in various organs and tissues, particularly the immune system and the liver. The review briefly summarizes the different models of humanized mice available for in vivo experiments. With a focus on lymphotropic, monocytotropic and hepatotropic viruses, we here discuss the current status and future prospects of these models for studying the pathogenesis of infectious diseases. Furthermore, they provide a powerful tool for the development of innovative therapies.
Collapse
Affiliation(s)
- Anne Cachat
- Virologie humaine, INSERM-U758, École Normale Supérieure, France
| | | | | | | | | |
Collapse
|
90
|
|
91
|
Abstract
The mononuclear phagocyte system (MPS) comprises monocytes, macrophages and dendritic cells. Tissue phagocytes share several cell surface markers, phagocytic capability and myeloid classification; however, the factors that regulate the differentiation, homeostasis and function of macrophages and dendritic cells remain largely unknown. The purpose of this manuscript is to review the tools that are currently available and those that are under development to study the origin and function of mononuclear phagocytes.
Collapse
|
92
|
Zhang L, Meissner E, Chen J, Su L. Current humanized mouse models for studying human immunology and HIV-1 immuno-pathogenesis. SCIENCE CHINA-LIFE SCIENCES 2010; 53:195-203. [PMID: 20596827 DOI: 10.1007/s11427-010-0059-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Accepted: 01/15/2010] [Indexed: 01/12/2023]
Abstract
A robust animal model for "hypothesis-testing/mechanistic" research in human immunology and immuno-pathology should meet the following criteria. First, it has well-studied hemato-lymphoid organs and target cells similar to those of humans. Second, the human pathogens establish infection and lead to relevant diseases. Third, it is genetically inbred and can be manipulated via genetic, immunological and pharmacological means. Many human-tropic pathogens such as HIV-1 fail to infect murine cells due to the blocks at multiple steps of their life cycle. The mouse with a reconstituted human immune system and other human target organs is a good candidate. A number of human-mouse chimeric models with human immune cells have been developed in the past 20 years, but most with only limited success due to the selective engraftment of xeno-reactive human T cells in hu-PBL-SCID mice or the lack of significant human immune responses in the SCID-hu Thy/Liv mouse. This review summarizes the current understanding of HIV-1 immuno-pathogenesis in human patients and in SIV-infected primate models. It also reviews the recent progress in the development of humanized mouse models with a functional human immune system, especially the recent progress in the immunodeficient mice that carry a defective gammaC gene. NOD/SCID/gammaC(-/-) (NOG or NSG) or the Rag2(-/-)gammaC(-/-) double knockout (DKO) mice, which lack NK as well as T and B cells (NTB-null mice), have been used to reconstitute a functional human immune system in central and peripheral lymphoid organs with human CD34(+) HSC. These NTB-hu HSC humanized models have been used to investigate HIV-1 infection, immuno-pathogenesis and therapeutic interventions. Such models, with further improvements, will contribute to study human immunology, human-tropic pathogens as well as human stem cell biology in the tissue development and function in vivo.
Collapse
Affiliation(s)
- LiGuo Zhang
- Key Laboratory of Immunity and Infection, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | | | | | | |
Collapse
|