1
|
Baltazar T, Jiang B, Moncayo A, Merola J, Albanna MZ, Saltzman WM, Pober JS. 3D bioprinting of an implantable xeno-free vascularized human skin graft. Bioeng Transl Med 2023; 8:e10324. [PMID: 36684084 PMCID: PMC9842062 DOI: 10.1002/btm2.10324] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 01/25/2023] Open
Abstract
Bioengineered tissues or organs produced using matrix proteins or components derived from xenogeneic sources pose risks of allergic responses, immune rejection, or even autoimmunity. Here, we report successful xeno-free isolation, expansion, and cryopreservation of human endothelial cells (EC), fibroblasts (FBs), pericytes (PCs), and keratinocytes (KCs). We further demonstrate the bioprinting of a human skin substitute with a dermal layer containing xeno-free cultured human EC, FBs, and PCs in a xeno-free bioink containing human collagen type I and fibronectin layered in a biocompatible polyglycolic acid mesh and subsequently seeded with xeno-free human KCs to form an epidermal layer. Following implantation of such bilayered skin grafts on the dorsum of immunodeficient mice, KCs form a mature stratified epidermis with rete ridge-like structures. The ECs and PCs form human EC-lined perfused microvessels within 2 weeks after implantation, preventing graft necrosis, and eliciting further perfusion of the graft by angiogenic host microvessels. As proof-of-concept, we generated 12 individual grafts using a single donor of all four cell types. In summary, we describe the fabrication of a bioprinted vascularized bilayered skin substitute under completely xeno-free culture conditions demonstrating feasibility of a xeno-free approach to complex tissue engineering.
Collapse
Affiliation(s)
- Tania Baltazar
- Department of Immunobiology, Yale School of Medicine New Haven Connecticut USA
| | - Bo Jiang
- Department of Surgery Yale University School of Medicine New Haven Connecticut USA
- Department of Vascular Surgery The First Hospital of China Medical University Shenyang China
| | - Alejandra Moncayo
- Department of Chronic Disease Epidemiology Yale University School of Public Health New Haven Connecticut USA
- College of Medicine SUNY Downstate Health Sciences University Brooklyn New York USA
| | - Jonathan Merola
- Department of Surgery Yale University School of Medicine New Haven Connecticut USA
- Department of Surgery Columbia University Medical Center New York New York USA
| | - Mohammad Z Albanna
- Humabiologics Inc Phoenix Arizona USA
- Department of General Surgery Atrium Health Wake Forest Baptist Winston-Salem North Carolina USA
| | - W Mark Saltzman
- Department of Biomedical Engineering Yale University New Haven Connecticut USA
| | - Jordan S Pober
- Department of Immunobiology, Yale School of Medicine New Haven Connecticut USA
| |
Collapse
|
2
|
Cristóbal L, Asúnsolo Á, Sánchez J, Ortega MA, Álvarez-Mon M, García-Honduvilla N, Buján J, Maldonado AA. Mouse Models for Human Skin Transplantation: A Systematic Review. Cells Tissues Organs 2021; 210:250-259. [PMID: 34521089 DOI: 10.1159/000516154] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 03/22/2021] [Indexed: 11/19/2022] Open
Abstract
Immunodeficient mouse models with human skin xenografts have been developed in the past decades to study different conditions of the skin. Features such as follow-up period and size of the graft are of different relevance depending on the purpose of an investigation. The aim of this study is to analyze the different mouse models grafted with human skin. A systematic review of the literature was performed in line with the PRISMA statement using MEDLINE/PubMed databases from January 1970 to June 2020. Articles describing human skin grafted onto mice were included. Animal models other than mice, skin substitutes, bioengineered skin, postmortem or fetal skin, and duplicated studies were excluded. The mouse strain, origin of human skin, graft dimensions, follow-up of the skin graft, and goals of the study were analyzed. Ninety-one models were included in the final review. Five different applications were found: physiology of the skin (25 models, mean human skin graft size 1.43 cm2 and follow-up 72.92 days), immunology and graft rejection (17 models, mean human skin graft size 1.34 cm2 and follow-up 86 days), carcinogenesis (9 models, mean human skin graft size 1.98 cm2 and follow-up 253 days), skin diseases (25 models, mean human skin graft size 1.55 cm2 and follow-up 86.48 days), and would healing/scars (15 models, mean human skin graft size 2.54 cm2 and follow-up 129 days). The follow-up period was longer in carcinogenesis models (253 ± 233.73 days), and the skin graft size was bigger in wound healing applications (2.54 ± 3.08 cm2). Depending on the research application, different models are suggested. Careful consideration regarding graft size, follow-up, immunosuppression, and costs should be analyzed and compared before choosing any of these mouse models. To our knowledge, this is the first systematic review of mouse models with human skin transplantation.
Collapse
Affiliation(s)
- Lara Cristóbal
- Department of Plastic Surgery and Burn Unit, University Hospital of Getafe, Madrid, Spain.,Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain.,Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain.,Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain.,Department of Epidemiology and Biostatistics, Graduate School of Public Health and Health Policy, The City University of New York, New York, New York, USA
| | - Jorge Sánchez
- Department of Plastic Surgery and Burn Unit, University Hospital of Getafe, Madrid, Spain,
| | - Miguel A Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain.,Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain.,Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain.,Immune System Diseases-Rheumatology, Oncology Service and Internal Medicine, CIBEREHD, University Hospital Príncipe de Asturias, Alcalá de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain.,Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Julia Buján
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain.,Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Andrés A Maldonado
- Department of Plastic Surgery and Burn Unit, University Hospital of Getafe, Madrid, Spain.,Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain.,Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain.,Department for Plastic, Hand and Reconstructive Surgery, BG Trauma Center Frankfurt am Main, Academic Hospital of the Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
3
|
Hopp CS, Kanatani S, Archer NK, Miller RJ, Liu H, Chiou KK, Miller LS, Sinnis P. Comparative intravital imaging of human and rodent malaria sporozoites reveals the skin is not a species-specific barrier. EMBO Mol Med 2021; 13:e11796. [PMID: 33750026 PMCID: PMC8033530 DOI: 10.15252/emmm.201911796] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 01/09/2023] Open
Abstract
Malaria infection starts with the injection of Plasmodium sporozoites into the host’s skin. Sporozoites are motile and move in the skin to find and enter blood vessels to be carried to the liver. Here, we present the first characterization of P. falciparum sporozoites in vivo, analyzing their motility in mouse skin and human skin xenografts and comparing their motility to two rodent malaria species. These data suggest that in contrast to the liver and blood stages, the skin is not a species‐specific barrier for Plasmodium. Indeed, P. falciparum sporozoites enter blood vessels in mouse skin at similar rates to the rodent malaria parasites. Furthermore, we demonstrate that antibodies targeting sporozoites significantly impact the motility of P. falciparum sporozoites in mouse skin. Though the sporozoite stage is a validated vaccine target, vaccine trials have been hampered by the lack of good animal models for human malaria parasites. Pre‐clinical screening of next‐generation vaccines would be significantly aided by the in vivo platform we describe here, expediting down‐selection of candidates prior to human vaccine trials.
Collapse
Affiliation(s)
- Christine S Hopp
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Sachie Kanatani
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevin K Chiou
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Peelen DM, Hoogduijn MJ, Hesselink DA, Baan CC. Advanced in vitro Research Models to Study the Role of Endothelial Cells in Solid Organ Transplantation. Front Immunol 2021; 12:607953. [PMID: 33664744 PMCID: PMC7921837 DOI: 10.3389/fimmu.2021.607953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/21/2021] [Indexed: 12/26/2022] Open
Abstract
The endothelium plays a key role in acute and chronic rejection of solid organ transplants. During both processes the endothelium is damaged often with major consequences for organ function. Also, endothelial cells (EC) have antigen-presenting properties and can in this manner initiate and enhance alloreactive immune responses. For decades, knowledge about these roles of EC have been obtained by studying both in vitro and in vivo models. These experimental models poorly imitate the immune response in patients and might explain why the discovery and development of agents that control EC responses is hampered. In recent years, various innovative human 3D in vitro models mimicking in vivo organ structure and function have been developed. These models will extend the knowledge about the diverse roles of EC in allograft rejection and will hopefully lead to discoveries of new targets that are involved in the interactions between the donor organ EC and the recipient's immune system. Moreover, these models can be used to gain a better insight in the mode of action of the currently prescribed immunosuppression and will enhance the development of novel therapeutics aiming to reduce allograft rejection and prolong graft survival.
Collapse
Affiliation(s)
- Daphne M Peelen
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Martin J Hoogduijn
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dennis A Hesselink
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Carla C Baan
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
5
|
Successful Regulatory T Cell-Based Therapy Relies on Inhibition of T Cell Effector Function and Enrichment of FOXP3+ Cells in a Humanized Mouse Model of Skin Inflammation. J Immunol Res 2020; 2020:7680131. [PMID: 32509883 PMCID: PMC7244960 DOI: 10.1155/2020/7680131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/19/2020] [Accepted: 01/30/2020] [Indexed: 11/17/2022] Open
Abstract
Background Recent clinical trials using regulatory T cells (Treg) support the therapeutic potential of Treg-based therapy in transplantation and autoinflammatory diseases. Despite these clinical successes, the effect of Treg on inflamed tissues, as well as their impact on immune effector function in vivo, is poorly understood. Therefore, we here evaluated the effect of human Treg injection on cutaneous inflammatory processes in vivo using a humanized mouse model of human skin inflammation (huPBL-SCID-huSkin). Methods SCID beige mice were transplanted with human skin followed by intraperitoneal (IP) injection of 20‐40 × 106 allogeneic human PBMCs. This typically results in human skin inflammation as indicated by epidermal thickening (hyperkeratosis) and changes in dermal inflammatory markers such as the antimicrobial peptide hBD2 and epidermal barrier cytokeratins K10 and K16, as well as T cell infiltration in the dermis. Ex vivo-expanded human Treg were infused intraperitoneally. Human cutaneous inflammation and systemic immune responses were analysed by immunohistochemistry and flow cytometry. Results We confirmed that human Treg injection inhibits skin inflammation and the influx of effector T cells. As a novel finding, we demonstrate that human Treg injection led to a reduction of IL-17-secreting cells while promoting a relative increase in immunosuppressive FOXP3+ Treg in the human skin, indicating active immune regulation in controlling the local proinflammatory response. Consistent with the local control (skin), systemically (splenocytes), we observed that Treg injection led to lower frequencies of IFNγ and IL-17A-expressing human T cells, while a trend towards enrichment of FOXP3+ Treg was observed. Conclusion Taken together, we demonstrate that inhibition of skin inflammation by Treg infusion, next to a reduction of infiltrating effector T cells, is mediated by restoring both the local and systemic balance between cytokine-producing effector T cells and immunoregulatory T cells. This work furthers our understanding of Treg-based immunotherapy.
Collapse
|
6
|
Lim S, Kirkiles-Smith NC, Pober JS, Bothwell ALM, Choi JM. Regulation of human T cell responses by dNP2-ctCTLA-4 inhibits human skin and microvessel graft rejection. Biomaterials 2018; 183:128-138. [PMID: 30165256 DOI: 10.1016/j.biomaterials.2018.08.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/14/2018] [Accepted: 08/20/2018] [Indexed: 12/30/2022]
Abstract
Manipulation of human T cell functioning by delivery of macromolecules such as DNA, RNA, or protein is limited, unless the human T cells have been stimulated or electropermeabilized. To achieve successful adaptation and survival of a grafted organ, the alloreactive T cells that induce graft rejection must be regulated. Corticosteroids, calcineurin inhibitors, and mTOR inhibitors, which are systemic immunosuppressants, are currently used for transplantation, with significant side effects. In this study, we demonstrated that a cell-permeable peptide (CPP), dNP2, could efficiently deliver proteins into human CD4 and CD8 T cells. We confirmed regulatory functioning of the cytoplasmic domain of CTLA-4 conjugated with dNP2 (dNP2-ctCTLA-4) in human T cell activation, proliferation, and chemokine receptor expression. We utilized a human skin allograft system in SCID/beige mice to examine whether dNP2-ctCTLA-4 could inhibit allograft rejection by controlling T cell responses. The grafted skin tissue inflammation, allogeneic T cell infiltration, and blood cytokine level was markedly reduced by dNP2-ctCTLA-4, resulting in successful transplantation. In addition, it also inhibited T cell alloresponses against microvessels formed form Bcl-2-transduced human umbilical vein endothelial cells implanted into Balb/c Rag1-/-/IL-2Rγ-/- double knockout (DKO) mice, assessed as reduced T cell infiltration and granzyme B expression. These results collectively suggest that dNP2 peptide conjugation offers a valuable tool for delivering macromolecules like proteins into human T cells, and dNP2-ctCTLA-4 is a novel agent that shows potential in controlling human T cell responses to allow successful adaptation of grafted tissues.
Collapse
Affiliation(s)
- Sangho Lim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea; Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Nancy C Kirkiles-Smith
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Alfred L M Bothwell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea; Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea.
| |
Collapse
|
7
|
Landman S, de Oliveira VL, van Erp PEJ, Fasse E, Bauland SCG, Joosten I, Koenen HJPM. Intradermal injection of low dose human regulatory T cells inhibits skin inflammation in a humanized mouse model. Sci Rep 2018; 8:10044. [PMID: 29968819 PMCID: PMC6030170 DOI: 10.1038/s41598-018-28346-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 06/11/2018] [Indexed: 12/31/2022] Open
Abstract
Recent regulatory T cell (Treg) based clinical trials support their therapeutic potential in transplantation and auto-inflammatory diseases. However, large numbers of Treg are needed to accomplish therapeutic efficacy. Local injection at the site of inflammation (targeted delivery) may lower the numbers needed for therapy. We evaluated if local delivery of low numbers of human Treg by intradermal injection was able to prevent skin inflammation, using the humanized mouse huPBL-SCID-huSkin allograft model. A dose of only 1 × 105 freshly isolated, non expanded Treg injected intradermally in close proximity to the transplanted human skin prevented inflammation of the grafted tissue induced by 4 × 107 IP injected human allogeneic PBMCs, (ratio Treg:PBMC = 1:400), as indicated by the inhibition of epidermal thickening, sustained Keratin-10 expression, the absence of Keratin-16 up regulation and prevention of human CD3+ T cell influx. A concomitant reduction of human T cells was observed in lymph nodes and spleen of the mice. Injection of Treg at the contralateral side was also shown to inhibit skin inflammation, suggesting that the inflammatory response was regulated both locally and systemically. In conclusion, local application of Treg may be an attractive way to suppress inflammation in vivo without the need for prior ex vivo expansion.
Collapse
Affiliation(s)
- Sija Landman
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | - Vivian L de Oliveira
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | - Piet E J van Erp
- Radboud university medical center, department of Dermatology, Nijmegen, The Netherlands
| | - Esther Fasse
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | | | - Irma Joosten
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands.
| |
Collapse
|
8
|
Liu R, Merola J, Manes TD, Qin L, Tietjen GT, López-Giráldez F, Broecker V, Fang C, Xie C, Chen PM, Kirkiles-Smith NC, Jane-Wit D, Pober JS. Interferon-γ converts human microvascular pericytes into negative regulators of alloimmunity through induction of indoleamine 2,3-dioxygenase 1. JCI Insight 2018. [PMID: 29515027 DOI: 10.1172/jci.insight.97881] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Early acute rejection of human allografts is mediated by circulating alloreactive host effector memory T cells (TEM). TEM infiltration typically occurs across graft postcapillary venules and involves sequential interactions with graft-derived endothelial cells (ECs) and pericytes (PCs). While the role of ECs in allograft rejection has been extensively studied, contributions of PCs to this process are largely unknown. This study aimed to characterize the effects and mechanisms of interactions between human PCs and allogeneic TEM. We report that unstimulated PCs, like ECs, can directly present alloantigen to TEM, but while IFN-γ-activated ECs (γ-ECs) show increased ability to stimulate alloreactive T cells, IFN-γ-activated PCs (γ-PCs) instead suppress TEM proliferation but not cytokine production or signaling. RNA sequencing analysis of PCs, γ-PCs, ECs, and γ-ECs reveal induction of indoleamine 2,3-dioxygenase 1 (IDO1) in γ-PCs to significantly higher levels than in γ-ECs that correlates with tryptophan depletion in vitro. Consistently, shRNA knockdown of IDO1 markedly reduces γ-PC-mediated immunoregulatory effects. Furthermore, human PCs express IDO1 in a skin allograft rejection humanized mouse model and in human renal allografts with acute T cell-mediated rejection. We conclude that immunosuppressive properties of human PCs are not intrinsic but instead result from IFN-γ-induced IDO1-mediated tryptophan depletion.
Collapse
Affiliation(s)
| | - Jonathan Merola
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Lingfeng Qin
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Gregory T Tietjen
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Verena Broecker
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Caodi Fang
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | | | | | - Dan Jane-Wit
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
9
|
Kenney LL, Shultz LD, Greiner DL, Brehm MA. Humanized Mouse Models for Transplant Immunology. Am J Transplant 2016; 16:389-97. [PMID: 26588186 PMCID: PMC5283075 DOI: 10.1111/ajt.13520] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/02/2015] [Accepted: 09/04/2015] [Indexed: 01/25/2023]
Abstract
Our understanding of the molecular pathways that control immune responses, particularly immunomodulatory molecules that control the extent and duration of an immune response, have led to new approaches in the field of transplantation immunology to induce allograft survival. These molecular pathways are being defined precisely in murine models and translated into clinical practice; however, many of the newly available drugs are human-specific reagents. Furthermore, many species-specific differences exist between mouse and human immune systems. Recent advances in the development of humanized mice, namely, immunodeficient mice engrafted with functional human immune systems, have led to the availability of a small animal model for the study of human immune responses. Humanized mice represent an important preclinical model system for evaluation of new drugs and identification of the mechanisms underlying human allograft rejection without putting patients at risk. This review highlights recent advances in the development of humanized mice and their use as preclinical models for the study of human allograft responses.
Collapse
Affiliation(s)
- Laurie L Kenney
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605
| | | | - Dale L Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605,Corresponding Author: Dale L. Greiner, PhD, University of Massachusetts Medical School, 368 Plantation Street, AS7-2051, Worcester, MA 01605, Office: 508-856-1911, Fax: 508-856-4093,
| | - Michael A. Brehm
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605
| |
Collapse
|
10
|
Safinia N, Becker PD, Vaikunthanathan T, Xiao F, Lechler R, Lombardi G. Humanized Mice as Preclinical Models in Transplantation. Methods Mol Biol 2016; 1371:177-196. [PMID: 26530801 DOI: 10.1007/978-1-4939-3139-2_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Animal models have been instrumental in our understanding of the mechanisms of rejection and the testing of novel treatment options in the context of transplantation. We have now entered an exciting era with research on humanized mice driving advances in translational studies and in our understanding of the function of human cells in response to pathogens and cancer as well as the recognition of human allogeneic tissues in vivo. In this chapter we provide a historical overview of humanized mouse models of transplantation to date, outlining the distinct strains and share our experiences in the study of human transplantation immunology.
Collapse
Affiliation(s)
- N Safinia
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK
| | - P D Becker
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK
| | - T Vaikunthanathan
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK
| | - F Xiao
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK
| | - R Lechler
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK
| | - G Lombardi
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK.
| |
Collapse
|
11
|
Maldonado AA, Cristóbal L, Martín-López J, Mallén M, García-Honduvilla N, Buján J. A novel model of human skin pressure ulcers in mice. PLoS One 2014; 9:e109003. [PMID: 25310568 PMCID: PMC4195607 DOI: 10.1371/journal.pone.0109003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/03/2014] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Pressure ulcers are a prevalent health problem in today's society. The shortage of suitable animal models limits our understanding and our ability to develop new therapies. This study aims to report on the development of a novel and reproducible human skin pressure ulcer model in mice. MATERIAL AND METHODS Male non-obese, diabetic, severe combined immunodeficiency mice (n = 22) were engrafted with human skin. A full-thickness skin graft was placed onto 4×3 cm wounds created on the dorsal skin of the mice. Two groups with permanent grafts were studied after 60 days. The control group (n = 6) was focused on the process of engraftment. Evaluations were conducted with photographic assessment, histological analysis and fluorescence in situ hybridization (FISH) techniques. The pressure ulcer group (n = 12) was created using a compression device. A pressure of 150 mmHg for 8 h, with a total of three cycles of compression-release was exerted. Evaluations were conducted with photographic assessment and histological analysis. RESULTS Skin grafts in the control group took successfully, as shown by visual assessment, FISH techniques and histological analysis. Pressure ulcers in the second group showed full-thickness skin loss with damage and necrosis of all the epidermal and dermal layers (ulcer stage III) in all cases. Complete repair occurred after 40 days. CONCLUSIONS An inexpensive, reproducible human skin pressure ulcer model has been developed. This novel model will facilitate the development of new clinically relevant therapeutic strategies that can be tested directly on human skin.
Collapse
Affiliation(s)
- Andrés A. Maldonado
- Department of Plastic and Reconstructive Surgery and Burn Unit, University Hospital of Getafe, Madrid, Spain
- * E-mail:
| | - Lara Cristóbal
- Department of Plastic and Reconstructive Surgery and Burn Unit, University Hospital of Getafe, Madrid, Spain
| | - Javier Martín-López
- Department of Pathology, University Hospital of Puerta de Hierro, Madrid, Spain
| | - Mar Mallén
- Department of Genetics, University Hospital Central de la Defensa, Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medical Specialties, Faculty of Medicine, University of Alcalá, Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Julia Buján
- Department of Medical Specialties, Faculty of Medicine, University of Alcalá, Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
12
|
Melican K, Aubey F, Duménil G. Humanized mouse model to study bacterial infections targeting the microvasculature. J Vis Exp 2014. [PMID: 24747976 PMCID: PMC4161007 DOI: 10.3791/51134] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Neisseria meningitidis causes a severe, frequently fatal sepsis when it enters the human blood stream. Infection leads to extensive damage of the blood vessels resulting in vascular leak, the development of purpuric rashes and eventual tissue necrosis. Studying the pathogenesis of this infection was previously limited by the human specificity of the bacteria, which makes in vivo models difficult. In this protocol, we describe a humanized model for this infection in which human skin, containing dermal microvessels, is grafted onto immunocompromised mice. These vessels anastomose with the mouse circulation while maintaining their human characteristics. Once introduced into this model, N. meningitidis adhere exclusively to the human vessels, resulting in extensive vascular damage, inflammation and in some cases the development of purpuric rash. This protocol describes the grafting, infection and evaluation steps of this model in the context of N. meningitidis infection. The technique may be applied to numerous human specific pathogens that infect the blood stream.
Collapse
Affiliation(s)
- Keira Melican
- INSERM U970, Paris Cardiovascular Research Centre; Faculté de Médecine Paris Descartes, Université Paris Descartes
| | - Flore Aubey
- INSERM U970, Paris Cardiovascular Research Centre; Faculté de Médecine Paris Descartes, Université Paris Descartes
| | - Guillaume Duménil
- INSERM U970, Paris Cardiovascular Research Centre; Faculté de Médecine Paris Descartes, Université Paris Descartes;
| |
Collapse
|
13
|
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
|
14
|
Melican K, Michea Veloso P, Martin T, Bruneval P, Duménil G. Adhesion of Neisseria meningitidis to dermal vessels leads to local vascular damage and purpura in a humanized mouse model. PLoS Pathog 2013; 9:e1003139. [PMID: 23359320 PMCID: PMC3554624 DOI: 10.1371/journal.ppat.1003139] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 12/03/2012] [Indexed: 12/12/2022] Open
Abstract
Septic shock caused by Neisseria meningitidis is typically rapidly evolving and often fatal despite antibiotic therapy. Further understanding of the mechanisms underlying the disease is necessary to reduce fatality rates. Postmortem samples from the characteristic purpuric rashes of the infection show bacterial aggregates in close association with microvessel endothelium but the species specificity of N. meningitidis has previously hindered the development of an in vivo model to study the role of adhesion on disease progression. Here we introduced human dermal microvessels into SCID/Beige mice by xenografting human skin. Bacteria injected intravenously exclusively associated with the human vessel endothelium in the skin graft. Infection was accompanied by a potent inflammatory response with the secretion of human inflammatory cytokines and recruitment of inflammatory cells. Importantly, infection also led to local vascular damage with hemostasis, thrombosis, vascular leakage and finally purpura in the grafted skin, replicating the clinical presentation for the first time in an animal model. The adhesive properties of the type IV pili of N. meningitidis were found to be the main mediator of association with the dermal microvessels in vivo. Bacterial mutants with altered type IV pili function also did not trigger inflammation or lead to vascular damage. This work demonstrates that local type IV pili mediated adhesion of N. meningitidis to the vascular wall, as opposed to circulating bacteria, determines vascular dysfunction in meningococcemia.
Collapse
Affiliation(s)
- Keira Melican
- INSERM, U970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Faculté de Médecine Paris Descartes, Paris, France
| | - Paula Michea Veloso
- INSERM, U970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Faculté de Médecine Paris Descartes, Paris, France
| | - Tiffany Martin
- INSERM, U970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Faculté de Médecine Paris Descartes, Paris, France
| | - Patrick Bruneval
- INSERM, U970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Faculté de Médecine Paris Descartes, Paris, France
- AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Guillaume Duménil
- INSERM, U970, Paris Cardiovascular Research Center, Paris, France
- Université Paris Descartes, Faculté de Médecine Paris Descartes, Paris, France
| |
Collapse
|
15
|
Unger WWJ, Pearson T, Abreu JRF, Laban S, van der Slik AR, der Kracht SMV, Kester MGD, Serreze DV, Shultz LD, Griffioen M, Drijfhout JW, Greiner DL, Roep BO. Islet-specific CTL cloned from a type 1 diabetes patient cause beta-cell destruction after engraftment into HLA-A2 transgenic NOD/scid/IL2RG null mice. PLoS One 2012; 7:e49213. [PMID: 23155466 PMCID: PMC3498321 DOI: 10.1371/journal.pone.0049213] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 10/05/2012] [Indexed: 11/21/2022] Open
Abstract
Despite increasing evidence that autoreactive CD8 T-cells are involved in both the initiation of type 1 diabetes (T1D) and the destruction of beta-cells, direct evidence for their destructive role in-vivo is lacking. To address a destructive role for autoreactive CD8 T-cells in human disease, we assessed the pathogenicity of a CD8 T-cell clone derived from a T1D donor and specific for an HLA-A2-restricted epitope of islet-specific glucose-6-phosphatase catalytic-subunit related protein (IGRP). HLA-A2/IGRP tetramer staining revealed a higher frequency of IGRP-specific CD8 T-cells in the peripheral blood of recent onset human individuals than of healthy donors. IGRP265–273-specific CD8 T-cells that were cloned from the peripheral blood of a recent onset T1D individual were shown to secrete IFNγ and Granzyme B after antigen-specific activation and lyse HLA-A2-expressing murine islets in-vitro. Lytic capacity was also demonstrated in-vivo by specific killing of peptide-pulsed target cells. Using the HLA-A2 NOD-scid IL2rγnull mouse model, HLA-A2-restricted IGRP-specific CD8 T-cells induced a destructive insulitis. Together, this is the first evidence that human HLA-restricted autoreactive CD8 T-cells target HLA-expressing beta-cells in-vivo, demonstrating the translational value of humanized mice to study mechanisms of disease and therapeutic intervention strategies.
Collapse
Affiliation(s)
- Wendy W. J. Unger
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Todd Pearson
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Joana R. F. Abreu
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Sandra Laban
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Arno R. van der Slik
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Sacha Mulder-van der Kracht
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Michel G. D. Kester
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dave V. Serreze
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Wouter Drijfhout
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Dale L. Greiner
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Bart O. Roep
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
16
|
de Oliveira VL, Keijsers RRMC, van de Kerkhof PCM, Seyger MMB, Fasse E, Svensson L, Latta M, Norsgaard H, Labuda T, Hupkens P, van Erp PEJ, Joosten I, Koenen HJPM. Humanized mouse model of skin inflammation is characterized by disturbed keratinocyte differentiation and influx of IL-17A producing T cells. PLoS One 2012; 7:e45509. [PMID: 23094018 PMCID: PMC3477148 DOI: 10.1371/journal.pone.0045509] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 08/21/2012] [Indexed: 12/14/2022] Open
Abstract
Humanized mouse models offer a challenging possibility to study human cell function in vivo. In the huPBL-SCID-huSkin allograft model human skin is transplanted onto immunodeficient mice and allowed to heal. Thereafter allogeneic human peripheral blood mononuclear cells are infused intra peritoneally to induce T cell mediated inflammation and microvessel destruction of the human skin. This model has great potential for in vivo study of human immune cells in (skin) inflammatory processes and for preclinical screening of systemically administered immunomodulating agents. Here we studied the inflammatory skin response of human keratinocytes and human T cells and the concomitant systemic human T cell response. As new findings in the inflamed human skin of the huPBL-SCID-huSkin model we here identified: 1. Parameters of dermal pathology that enable precise quantification of the local skin inflammatory response exemplified by acanthosis, increased expression of human β-defensin-2, Elafin, K16, Ki67 and reduced expression of K10 by microscopy and immunohistochemistry. 2. Induction of human cytokines and chemokines using quantitative real-time PCR. 3. Influx of inflammation associated IL-17A-producing human CD4+ and CD8+ T cells as well as immunoregulatory CD4+Foxp3+ cells using immunohistochemistry and -fluorescence, suggesting that active immune regulation is taking place locally in the inflamed skin. 4. Systemic responses that revealed activated and proliferating human CD4+ and CD8+ T cells that acquired homing marker expression of CD62L and CLA. Finally, we demonstrated the value of the newly identified parameters by showing significant changes upon systemic treatment with the T cell inhibitory agents cyclosporine-A and rapamycin. In summary, here we equipped the huPBL-SCID-huSkin humanized mouse model with relevant tools not only to quantify the inflammatory dermal response, but also to monitor the peripheral immune status. This combined approach will gain our understanding of the dermal immunopathology in humans and benefit the development of novel therapeutics for controlling inflammatory skin diseases.
Collapse
Affiliation(s)
- Vivian L. de Oliveira
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Romy R. M. C. Keijsers
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- Dermatology Department, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | - Marieke M. B. Seyger
- Dermatology Department, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Esther Fasse
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Lars Svensson
- Department of Disease Pharmacology, LEO Pharma, Ballerup, Denmark
| | - Markus Latta
- Department of Disease Pharmacology, LEO Pharma, Ballerup, Denmark
| | - Hanne Norsgaard
- Department of Molecular Biomedicine, LEO Pharma, Ballerup, Denmark
| | - Tord Labuda
- Department of Molecular Biomedicine, LEO Pharma, Ballerup, Denmark
| | - Pieter Hupkens
- Department of Plastic Surgery Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Piet E. J. van Erp
- Dermatology Department, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Irma Joosten
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Hans J. P. M. Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
17
|
Hossain M, Qadri SM, Liu L. Inhibition of nitric oxide synthesis enhances leukocyte rolling and adhesion in human microvasculature. JOURNAL OF INFLAMMATION-LONDON 2012; 9:28. [PMID: 22812684 PMCID: PMC3414823 DOI: 10.1186/1476-9255-9-28] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 07/19/2012] [Indexed: 02/08/2023]
Abstract
Background Nitric oxide (NO) is a multifunctional signaling molecule that regulates important cellular events in inflammation including leukocyte recruitment. Previous studies have shown that pharmacological inhibition of NO synthesis induces leukocyte recruitment in various in vitro and animal models. However, it is not known whether NO modulation has similar effects on leukocyte-endothelial cell interactions within the human microvasculature. The present study explored the effect of systemic L-NAME treatment on leukocyte recruitment in the SCID-hu mouse model. Methods Human skin xenografts were transplanted in SCID mice to study human leukocyte dynamics in human vasculature. Early events of human leukocyte recruitment in human vasculature were studied using intravital microscopy. NO synthesis was pharmacologically inhibited using NG-nitro-L-arginine methyl ester (L-NAME). Immunohistochemical analysis was performed to elucidate E-selectin expression in human xenograft skin. Human neutrophil-endothelial cell interactions were also studied in an in vitro flow chamber assay system. P- and E-selectin expression on cultured human umbilical vein endothelial cells (HUVECs) was measured using ELISA. Platelet-activating factor (PAF) synthesis was detected using a TLC-based assay. Results L-NAME treatment significantly enhanced the rolling and adhesion of human leukocytes to the human vasculature. Functional blocking of P- and E-selectins significantly inhibited rolling but not adhesion induced by inhibition of NO synthesis. Systemic L-NAME treatment enhanced E-selectin expression in human xenograft skin. L-NAME treatment significantly enhanced P- and E-selectin expression on HUVECs. L-NAME treatment did not significantly modify neutrophil rolling or adhesion to HUVECs indicating that L-NAME−induced subtle P- and E-selectin expression was insufficient to elicit dynamic neutrophil-HUVEC interactions in vitro. Moreover, synthesis of endothelial-derived PAF was not significantly modified by L-NAME treatment. These results point to the accelerated leukocyte recruitment in human vasculature following suppression of NO synthesis, effects that are mediated by P- and E-selectins. The findings are, however, not supported by the in vitro data. Conclusion Inhibition of endogenous NO triggers early events of human leukocyte recruitment in human vasculature, involving complex cellular or molecular mechanisms in addition to P- and E-selectin-mediated leukocyte rolling.
Collapse
Affiliation(s)
- Mokarram Hossain
- Department of Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
| | | | | |
Collapse
|
18
|
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: 2.2] [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
|
19
|
Nims RW, Sykes G, Cottrill K, Ikonomi P, Elmore E. Short tandem repeat profiling: part of an overall strategy for reducing the frequency of cell misidentification. In Vitro Cell Dev Biol Anim 2010; 46:811-9. [PMID: 20927602 PMCID: PMC2995877 DOI: 10.1007/s11626-010-9352-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 09/13/2010] [Indexed: 11/16/2022]
Abstract
The role of cell authentication in biomedical science has received considerable attention, especially within the past decade. This quality control attribute is now beginning to be given the emphasis it deserves by granting agencies and by scientific journals. Short tandem repeat (STR) profiling, one of a few DNA profiling technologies now available, is being proposed for routine identification (authentication) of human cell lines, stem cells, and tissues. The advantage of this technique over methods such as isoenzyme analysis, karyotyping, human leukocyte antigen typing, etc., is that STR profiling can establish identity to the individual level, provided that the appropriate number and types of loci are evaluated. To best employ this technology, a standardized protocol and a data-driven, quality-controlled, and publically searchable database will be necessary. This public STR database (currently under development) will enable investigators to rapidly authenticate human-based cultures to the individual from whom the cells were sourced. Use of similar approaches for non-human animal cells will require developing other suitable loci sets. While implementing STR analysis on a more routine basis should significantly reduce the frequency of cell misidentification, additional technologies may be needed as part of an overall authentication paradigm. For instance, isoenzyme analysis, PCR-based DNA amplification, and sequence-based barcoding methods enable rapid confirmation of a cell line's species of origin while screening against cross-contaminations, especially when the cells present are not recognized by the species-specific STR method. Karyotyping may also be needed as a supporting tool during establishment of an STR database. Finally, good cell culture practices must always remain a major component of any effort to reduce the frequency of cell misidentification.
Collapse
Affiliation(s)
- Raymond W. Nims
- RMC Pharmaceutical Solutions, Inc, 2150 Miller Drive, Suite A, Longmont, CO 80501 USA
| | - Greg Sykes
- ATCC, 10801 University Blvd., Manassas, VA 20110 USA
| | | | | | - Eugene Elmore
- Department of Radiation Oncology, University of California, Medical Sciences I, B146D, Irvine, CA 92697 USA
| |
Collapse
|
20
|
Abele-Ohl S, Leis M, Mahmoudian S, Weyand M, Stamminger T, Ensminger SM. Rag2-/- gamma-chain-/- mice as hosts for human vessel transplantation and allogeneic human leukocyte reconstitution. Transpl Immunol 2010; 23:59-64. [PMID: 20394817 DOI: 10.1016/j.trim.2010.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 03/30/2010] [Accepted: 04/05/2010] [Indexed: 11/18/2022]
Abstract
BACKGROUND Rodent models are a very helpful tool to investigate immunological mechanisms in allograft rejection. The aim of this study was to compare two different immunodeficient recipients in a humanized mouse model of arterial xenotransplantation in terms of reconstitution of the human immune system and rejection of the arterial graft. METHODS Side branches of human mammary artery were transplanted as infrarenal aortic interposition grafts into C.B-17-SCID beige and C57BL/6-Rag2(-/-)gammac(-/-) recipients. 7days after surgery mice were reconstituted with 5x10(7) human peripheral blood mononuclear cells (hu PBMCs) and 30days after reconstitution mice were sacrificed and histologic analysis was performed. Peripheral blood and splenocytes were investigated by FACS and ELISA analysis to ensure engraftment of human CD45(+) cells. RESULTS Transplant arteriosclerosis developed in non-PBMC-reconstituted C.B-17-SCID beige mice (intimal proliferation: 36.31+/-4.37%), but significantly less in C57BL/6-Rag2(-/-) gammac(-/-) recipients (intimal proliferation: 12.26+/-5.21%). After reconstitution with 5x10(7) unfractionated human PBMCs both mouse strains showed intima proliferation 30days after reconstitution (C.B-17-SCID beige: 28.49+/-7.95% and C57BL/6-Rag2(-/-) gammac(-/-): 44.58+/-11.08%). Whereas only very few human CD45(+) cells were found in mouse blood and spleen of C.B-17-SCID beige mice, C57BL/6-Rag2(-/-) gammac(-/-) mice revealed a reliable reconstitution. In addition, levels of human IgG and IgM within the peripheral blood were markedly higher in C57BL/6-Rag2(-/-) gammac(-/-) recipients. CONCLUSION In this study we can show, that the use of C57BL/6-Rag2(-/-) gammac(-/-) mice may be advantageous compared to C.B-17-SCID beige recipients in a humanized mouse model of vessel transplantation.
Collapse
Affiliation(s)
- Silke Abele-Ohl
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Krankenhausstrasse 12, 91054 Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
21
|
Lepus CM, Gibson TF, Gerber SA, Kawikova I, Szczepanik M, Hossain J, Ablamunits V, Kirkiles-Smith N, Herold KC, Donis RO, Bothwell AL, Pober JS, Harding MJ. Comparison of human fetal liver, umbilical cord blood, and adult blood hematopoietic stem cell engraftment in NOD-scid/gammac-/-, Balb/c-Rag1-/-gammac-/-, and C.B-17-scid/bg immunodeficient mice. Hum Immunol 2009; 70:790-802. [PMID: 19524633 DOI: 10.1016/j.humimm.2009.06.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Revised: 05/30/2009] [Accepted: 06/03/2009] [Indexed: 10/20/2022]
Abstract
Immunodeficient mice bearing components of a human immune system present a novel approach for studying human immune responses. We investigated the number, phenotype, developmental kinetics, and function of developing human immune cells following transfer of CD34(+) hematopoietic stem cell (HSC) preparations originating from second trimester human fetal liver (HFL), umbilical cord blood (UCB), or granulocyte colony-stimulating factor-mobilized adult blood (G-CSF-AB) delivered via intrahepatic injection into sublethally irradiated neonatal NOD-scid/gammac(-/-), Balb/c-Rag1(-/-)gammac(-/-), and C.B-17-scid/bg mice. HFL and UCB HSC provided the greatest number and breadth of developing cells. NOD-scid/gammac(-/-) and Balb/c-Rag1(-/-)gammac(-/-) harbored human B and dendritic cells as well as human platelets in peripheral blood, whereas NOD-scid/gammac(-/-) mice harbored higher levels of human T cells. NOD-scid/gammac(-/-) mice engrafted with HFL CD34(+) HSC demonstrated human immunological competence evidenced by white pulp expansion and increases in total human immunoglobulin following immunization with T-dependent antigens and delayed-type hypersensitivity-infiltrating leukocytes in response to antigenic challenge. In conclusion, we describe an encouraging base system for studying human hematopoietic lineage development and function utilizing human HFL or UCB HSC-engrafted NOD-scid/gammac(-/-) mice that is well suited for future studies toward the development of a fully competent humanized mouse model.
Collapse
Affiliation(s)
- Christin M Lepus
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06509, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Gilet J, Chang Y, Chenivesse C, Legendre B, Vorng H, Duez C, Wallaert B, Porte H, Senechal S, Tsicopoulos A. Role of CCL17 in the Generation of Cutaneous Inflammatory Reactions in Hu-PBMC-SCID Mice Grafted with Human Skin. J Invest Dermatol 2009; 129:879-90. [DOI: 10.1038/jid.2008.333] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
23
|
Development of a humanized mouse model to study the role of macrophages in allograft injury. Transplantation 2009; 87:189-97. [PMID: 19155972 DOI: 10.1097/tp.0b013e318192e05d] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Nearly half of all infiltrating leukocytes in rejecting human allografts are macrophages, yet, in comparison with T cells, much less is known about the contribution of this cell type to rejection. Our laboratory has previously described models of rejection of human skin or artery grafts in immunodeficient mouse hosts mediated by adoptively transferred allogeneic T cells. However, mature human monocyte/macrophages have consistently failed to engraft in these animals. Here, we describe the introduction of human CD68+ macrophages into irradiated immunodeficient mice by transplantation of enriched CD34+ hematopoietic stem-cells isolated from peripheral blood of G-colony-stimulating factor pretreated adults. METHODS We investigated strains of immunodeficient mice bearing human tissue grafts (skin and artery) inoculated with 1 x 10(6) human CD34+ adult hematopoietic stem cells, peripheral blood monuclear cells autologous to the CD34 donor, or both for human cell engraftment. RESULTS In the absence of T cells, CD68+ CD14+ macrophages infiltrate allogeneic human skin but produce little injury or thrombosis. Both responses are enhanced when combined with adoptive transfer of T cells autologous to the hematopoietic stem cells as exemplified by the induction of the macrophage activation marker CD163. CD68+ macrophages also infiltrate allogeneic arterial interposition grafts, producing intimal expansion and calcification in the absence of T cells. CONCLUSIONS These new models may be used to study the role of human macrophages in transplant rejection and other pathologies in vivo.
Collapse
|
24
|
Ren YR, Pan F, Parvez S, Fleig A, Chong CR, Xu J, Dang Y, Zhang J, Jiang H, Penner R, Liu JO. Clofazimine inhibits human Kv1.3 potassium channel by perturbing calcium oscillation in T lymphocytes. PLoS One 2008; 3:e4009. [PMID: 19104661 PMCID: PMC2602975 DOI: 10.1371/journal.pone.0004009] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2008] [Accepted: 11/08/2008] [Indexed: 01/03/2023] Open
Abstract
The Kv1.3 potassium channel plays an essential role in effector memory T cells and has been implicated in several important autoimmune diseases including multiple sclerosis, psoriasis and type 1 diabetes. A number of potent small molecule inhibitors of Kv1.3 channel have been reported, some of which were found to be effective in various animal models of autoimmune diseases. We report herein the identification of clofazimine, a known anti-mycobacterial drug, as a novel inhibitor of human Kv1.3. Clofazimine was initially identified as an inhibitor of intracellular T cell receptor-mediated signaling leading to the transcriptional activation of human interleukin-2 gene in T cells from a screen of the Johns Hopkins Drug Library. A systematic mechanistic deconvolution revealed that clofazimine selectively blocked the Kv1.3 channel activity, perturbing the oscillation frequency of the calcium-release activated calcium channel, which in turn led to the inhibition of the calcineurin-NFAT signaling pathway. These effects of clofazimine provide the first line of experimental evidence in support of a causal relationship between Kv1.3 and calcium oscillation in human T cells. Furthermore, clofazimine was found to be effective in blocking human T cell-mediated skin graft rejection in an animal model in vivo. Together, these results suggest that clofazimine is a promising immunomodulatory drug candidate for treating a variety of autoimmune disorders.
Collapse
Affiliation(s)
- Yunzhao R. Ren
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Program in Biochemistry, Cellular and Molecular Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Fan Pan
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Suhel Parvez
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, Hawaii, United States of America
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, Hawaii, United States of America
| | - Curtis R. Chong
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jing Xu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yongjun Dang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jin Zhang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Hongsi Jiang
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, Illinois, United States of America
| | - Reinhold Penner
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, Hawaii, United States of America
| | - Jun O. Liu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Program in Biochemistry, Cellular and Molecular Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
25
|
Rao DA, Eid RE, Qin L, Yi T, Kirkiles-Smith NC, Tellides G, Pober JS. Interleukin (IL)-1 promotes allogeneic T cell intimal infiltration and IL-17 production in a model of human artery rejection. ACTA ACUST UNITED AC 2008; 205:3145-58. [PMID: 19075290 PMCID: PMC2605225 DOI: 10.1084/jem.20081661] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Interleukin (IL) 1α produced by human endothelial cells (ECs), in response to tumor necrosis factor (TNF) or to co-culture with allogeneic T cells in a TNF-dependent manner, can augment the release of cytokines from alloreactive memory T cells in vitro. In a human–mouse chimeric model of artery allograft rejection, ECs lining the transplanted human arteries express IL-1α, and blocking IL-1 reduces the extent of human T cell infiltration into the artery intima and selectively inhibits IL-17 production by infiltrating T cells. In human skin grafts implanted on immunodeficient mice, administration of IL-17 is sufficient to induce mild inflammation. In cultured cells, IL-17 acts preferentially on vascular smooth muscle cells rather than ECs to enhance production of proinflammatory mediators, including IL-6, CXCL8, and CCL20. Neutralization of IL-17 does not reduce T cell infiltration into allogeneic human artery grafts, but markedly reduces IL-6, CXCL8, and CCL20 expression and selectively inhibits CCR6+ T cell accumulation in rejecting arteries. We conclude that graft-derived IL-1 can promote T cell intimal recruitment and IL-17 production during human artery allograft rejection, and suggest that targeting IL-1 in the perioperative transplant period may modulate host alloreactivity.
Collapse
Affiliation(s)
- Deepak A Rao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Vascular endothelial cells are an exposed target tissue for immune-mediated injury during graft-versus-host disease (GVHD). However, widespread endothelial death resulting in multi-organ failure similar to that in hyperacute solid-organ transplant rejection is not observed during GVHD. The rather mild endothelial injury seen in histological samples from affected skin biopsies contrasts with severe epithelial injury observed sometimes simultaneously. The elucidation of the mechanisms that influence endothelial susceptibility to immune-mediated injury would explain this paradox and may help to separate GVHD from the beneficial graft-versus-leukaemia effect. Transplant-associated microangiopathy, veno-occlusive disease and accelerated arteriosclerosis are vascular injury syndromes that occur after allogeneic stem-cell transplantation. Biomarkers are needed to identify individuals at risk of developing these complications. Treatments that have been found to be particularly effective for these specific endothelial injury syndromes need to be tested in larger clinical trials.
Collapse
|
27
|
Mathew JM, Blomberg B, Ricordi C, Esquenazi V, Miller J. Evaluation of the tolerogenic effects of donor bone marrow cells using a severe combined immunodeficient mouse-human islet transplant model. Hum Immunol 2008; 69:605-13. [PMID: 18703102 DOI: 10.1016/j.humimm.2008.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 07/02/2008] [Accepted: 07/17/2008] [Indexed: 10/21/2022]
Abstract
The immunoregulatory role of human donor bone marrow cells (DBMC) has been studied extensively in our laboratory using in vitro and ex vivo assays. However, new experimental systems that can overcome the limitations of tissue culture assays but with more clinical relevance than purely animal experimentation, needed to be generated. Therefore we have developed a new human peripheral blood lymphocyte (PBL) severe combined immunodeficient (SCID) mouse islet transplantation model without the occurrence of graft-versus-host disease (GvHD) and have used it to evaluate the tolerogenic effects of DBMC. Nonobese diabetogenic (NOD)-SCID mice were transplanted with human deceased donor islets and were reconstituted with human PBL (allogeneic to islets; denoted as recipient) with or without DBMC from the islet donor. It was observed that the most cellularly economical dose was 3000 islets per animal and that injection into the portal vein was better than implantation under the kidney capsule. Even though maximal lymphoid reconstitution was observed with 40-million fresh and anti-CD3 activated recipient PBL (conventional method), the mice developed severe graft GvHD. However, with the new method of reconstitution where animals were injected with 20-million anti-CD3-activated plus 40-million anti-donor-activated recipient PBL, no discernible GvHD was observed. More importantly, this latter method was associated with islet transplant rejection, which in turn could be abrogated by co-injection of the animals with DBMC. These in vivo results confirmed our previous in vitro observations that human DBMC have regulatory activity.
Collapse
Affiliation(s)
- James M Mathew
- Department of Surgery, Transplant Laboratories, Northwestern University, Chicago, IL, USA.
| | | | | | | | | |
Collapse
|
28
|
Zhang B, Duan Z, Zhao Y. Mouse models with human immunity and their application in biomedical research. J Cell Mol Med 2008; 13:1043-58. [PMID: 18419795 PMCID: PMC4496103 DOI: 10.1111/j.1582-4934.2008.00347.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Biomedical research in human beings is largely restricted to in vitro studies that lack complexity of a living organism. To overcome this limitation, humanized mouse models are developed based on immunodeficient characteristics of severe combined immunodeficiency (SCID) or recombination activating gene (Rag)(null) mice, which can accept xenografts. Peripheral constitution of human immunity in SCID or Rag(null) mice has been achieved by transplantation of mature human immune cells, foetal human thymus, bone marrow, liver tissues, lymph nodes or a combination of these, although efficiency needs to be improved. These mouse models with constituted human immunity (defined as humanized mice in the present text) have been widely used to investigate the basic principles of human immunobiology as well as complex pathomechanisms and potential therapies of human diseases. Here, elements of an ideal humanized mouse model are highlighted including genetic and non-genetic modification of recipient mice, transplantation strategies and proposals to improve engraftments. The applications of the humanized mice to study the development and response of human immune cells, human autoimmune diseases, virus infections, transplantation biology and tumour biology are reviewed as well.
Collapse
Affiliation(s)
- Baojun Zhang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | |
Collapse
|
29
|
Brands K, Colvin E, Williams LJ, Wang R, Lock RB, Tuch BE. Reduced immunogenicity of first-trimester human fetal pancreas. Diabetes 2008; 57:627-34. [PMID: 18065519 DOI: 10.2337/db07-0720a] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The use of human fetal pancreatic tissue may provide a potential source of transplantable beta-cells as a therapy for type 1 diabetes. Human fetal pancreas has a remarkable capacity to grow and differentiate in vivo and has been shown to reverse diabetes in rodents. However, it is known that human fetal pancreas obtained from the second trimester of gestation is immunogenic and is rejected after transplantation. Tissue obtained from earlier stages might prove to be immune privileged, as has been shown for other tissues. RESEARCH DESIGN AND METHODS In this study, we determined the immunogenicity of human fetal pancreatic tissue obtained from the first trimester of gestation in a humanized mouse model. A microarray study of immunoregulatory gene expression in first- and second-trimester human fetal pancreas was also undertaken. RESULTS The analysis of transplanted human fetal pancreata revealed a significantly decreased immunogenicity of the first-trimester tissue. The first-trimester grafts showed only limited cellular infiltration and contained numerous insulin-positive cells, whereas second-trimester tissue was completely infiltrated and rejected. Furthermore an analysis of immunoregulatory genes expressed in first- and second-trimester human fetal pancreas by microarray demonstrated the upregulation of several key immunoregulatory genes in the second-trimester tissue. This might account for the reduced immunogenicity of the younger tissue. CONCLUSIONS Our results provide the first indication that the use of first-trimester human fetal pancreas for transplantation might increase the survival of the grafts and might decrease the requirement for immunosuppressive drugs.
Collapse
Affiliation(s)
- Kerstin Brands
- Diabetes Transplant Unit, The Prince of Wales Hospital and The University of New South Wales, Sydney, New South Wales 2031, Australia
| | | | | | | | | | | |
Collapse
|
30
|
King M, Pearson T, Shultz LD, Leif J, Bottino R, Trucco M, Atkinson MA, Wasserfall C, Herold KC, Woodland RT, Schmidt MR, Woda BA, Thompson MJ, Rossini AA, Greiner DL. A new Hu-PBL model for the study of human islet alloreactivity based on NOD-scid mice bearing a targeted mutation in the IL-2 receptor gamma chain gene. Clin Immunol 2007; 126:303-14. [PMID: 18096436 DOI: 10.1016/j.clim.2007.11.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 11/07/2007] [Accepted: 11/12/2007] [Indexed: 11/25/2022]
Abstract
Immunodeficient NOD-scid mice bearing a targeted mutation in the IL2 receptor common gamma chain (Il2rgamma(null)) readily engraft with human stem cells. Here we analyzed human peripheral blood mononuclear cells (PBMC) for their ability to engraft NOD-scid Il2rgamma(null) mice and established engraftment kinetics, optimal cell dose, and the influence of injection route. Even at low PBMC input, NOD-scid Il2rgamma(null) mice reproducibly support high human PBMC engraftment that plateaus within 3-4 weeks. In contrast to previous stocks of immunodeficient mice, we observed low intra- and inter-donor variability of engraftment. NOD-scid Il2rgamma(null) mice rendered hyperglycemic by streptozotocin treatment return to normoglycemia following transplantation with human islets. Interestingly, these human islet grafts are rejected following injection of HLA-mismatched human PBMC as evidenced by return to hyperglycemia and loss of human C-peptide. These data suggest that humanized NOD-scid Il2rgamma(null) mice may represent an important surrogate for investigating in vivo mechanisms of human islet allograft rejection.
Collapse
Affiliation(s)
- Marie King
- Department of Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA 01655, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Shiao SL, Kirkiles-Smith NC, Shepherd BR, McNiff JM, Carr EJ, Pober JS. Human effector memory CD4+ T cells directly recognize allogeneic endothelial cells in vitro and in vivo. THE JOURNAL OF IMMUNOLOGY 2007; 179:4397-404. [PMID: 17878335 DOI: 10.4049/jimmunol.179.7.4397] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The frequency of circulating alloreactive human memory T cells correlates with allograft rejection. Memory T cells may be divided into effector memory (T(EM)) and central memory (T(CM)) cell subsets, but their specific roles in allograft rejection are unknown. We report that CD4+ T(EM) (CD45RO+ CCR7- CD62L-) can be adoptively transferred readily into C.B-17 SCID/bg mice and mediate the destruction of human endothelial cells (EC) in vascularized human skin grafts allogeneic to the T cell donor. In contrast, CD4+ T(CM) (CD45RO+ CCR7+ CD62L+) are inefficiently transferred and do not mediate EC injury. In vitro, CD4+ T(EM) secrete more IFN-gamma within 48 h in response to allogeneic ECs than do T(CM). In contrast, T(EM) and T(CM) secrete comparable amounts of IFN-gamma in response to allogeneic monocytes (Mo). In the same cultures, both T(EM) and T(CM) produce IL-2 and proliferate in response to IFN-gamma-treated allogeneic human EC or Mo, but T(CM) respond more vigorously in both assays. Blockade of LFA-3 strongly inhibits both IL-2 and IFN-gamma secretion by CD4+ T(EM) cultured with allogeneic EC but only minimally inhibits responses to allogeneic Mo. Blockade of CD80 and CD86 strongly inhibits IL-2 but not IFN-gamma production by in response to allogeneic EC or Mo. Transduction of EC to express B7-2 enhances allogeneic T(EM) production of IL-2 but not IFN-gamma. We conclude that human CD4+ T(EM) directly recognize and respond to allogeneic EC in vitro by secreting IFN-gamma and that this response depends on CD2 but not CD28. Consistent with EC activation of effector functions, human CD4+ T(EM) can mediate allogeneic EC injury in vivo.
Collapse
Affiliation(s)
- Stephen L Shiao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | |
Collapse
|
32
|
Moreno A, Pérignon JL, Morosan S, Mazier D, Benito A. Plasmodium falciparum-infected mice: more than a tour de force. Trends Parasitol 2007; 23:254-9. [PMID: 17434343 DOI: 10.1016/j.pt.2007.04.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2006] [Revised: 02/19/2007] [Accepted: 04/05/2007] [Indexed: 01/11/2023]
Abstract
Up until recently, the relevance of Plasmodium falciparum-infected humanized mice for malaria studies has been questioned because of the low percentage of mice in which the parasite develops. Advances in the generation of new immunodeficient mouse strains combined with the use of protocols that modulate the innate immune defenses of mice have facilitated the harvesting of exoerythrocytic and intraerythrocytic stages of the parasite. These results renew the hope of working with P. falciparum in a laboratory animal and indicate that the next challenge (i.e. a complete parasite cycle in the same mouse, including transmission to mosquito) could be reached in the future.
Collapse
Affiliation(s)
- Alicia Moreno
- Université Pierre & Marie Curie-Paris 6, UMR S511, Paris F-75013, France.
| | | | | | | | | |
Collapse
|
33
|
Shiao SL, McNiff JM, Masunaga T, Tamura K, Kubo K, Pober JS. Immunomodulatory properties of FK734, a humanized anti-CD28 monoclonal antibody with agonistic and antagonistic activities. Transplantation 2007; 83:304-13. [PMID: 17297405 DOI: 10.1097/01.tp.0000251426.46312.d5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND We describe immunomodulatory effects of FK734, a humanized version of a mouse anti-human CD28 mAb (clone TN228), in vitro and in a chimeric human-mouse model of allograft rejection. METHODS Cytokine production and proliferation were assessed in a mixed lymphocyte reaction containing FK734, human T cells, and endothelial cells or monocytes. FK734 was also administered to SCID mice engrafted with human skin and adoptively transferred with human peripheral blood mononuclear cells allogeneic to the skin graft. RESULTS In vitro, FK734 enhanced secretion of interleukin-2 and interferon-gamma as well as proliferation of CD4+ and CD8+ T cells stimulated by allogeneic human leukocyte antigen (HLA)-DR+ human umbilical vein endothelial cells (which lack B7 molecules and FcgammaRs) or by blood monocytes (which express low levels of B7 molecules and FcgammaRs) compared with control mAb, but these effects were significantly smaller than those provided by mAb 28.2, a stimulatory mouse anti-human CD28 mAb, at comparable concentrations. However, FK734 generally inhibited cytokine secretion and T cell proliferation in cocultures with human umbilical vein endothelial cells transduced to express CD86. In vivo using SCID/beige mice bearing human skin with adoptively transferred peripheral blood mononuclear cells, administration of FK734 protected human endothelial cell-lined microvessels, significantly but incompletely reducing endothelial cell injury and T cell infiltration into the graft one or two weeks later. CONCLUSIONS FK734 is a partial agonist of CD28 signaling that can reduce human T cell alloresponses in the presence of strong costimulation by B7 molecules in vitro and can reduce T cell-mediated skin allograft rejection in vivo.
Collapse
Affiliation(s)
- Stephen L Shiao
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
The culmination of decades of research on humanized mice is leading to advances in our understanding of human haematopoiesis, innate and adaptive immunity, autoimmunity, infectious diseases, cancer biology and regenerative medicine. In this Review, we discuss the development of these new generations of humanized mice, how they will facilitate translational research in several biomedical disciplines and approaches to overcome the remaining limitations of these models.
Collapse
Affiliation(s)
- Leonard D Shultz
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA.
| | | | | |
Collapse
|
35
|
Yamakuchi M, Kirkiles-Smith NC, Ferlito M, Cameron SJ, Bao C, Fox-Talbot K, Wasowska BA, Baldwin WM, Pober JS, Lowenstein CJ. Antibody to human leukocyte antigen triggers endothelial exocytosis. Proc Natl Acad Sci U S A 2007; 104:1301-6. [PMID: 17229850 PMCID: PMC1783109 DOI: 10.1073/pnas.0602035104] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Indexed: 11/18/2022] Open
Abstract
Although antibodies to HLA play a role in the pathogenesis of diseases processes such as rejection of transplanted organs, the precise mechanisms by which antibodies cause tissue injury are not completely understood. We hypothesized that antibodies to host tissues cause inflammation in part by activating endothelial exocytosis of granules that contain prothrombotic mediators such as von Willebrand Factor (VWF) and proinflammatory mediators such as P-selectin. To test this hypothesis, we treated human endothelial cells with murine monoclonal antibody W6/32 to HLA class I and then measured exocytosis by the release of VWF and the externalization of P-selectin. Antibody to HLA activates endothelial exocytosis in a dose-dependent manner over time. The biologically active complement split product, C5a, adds a slight but significant increase to antibody induction of exocytosis. Antibody to HLA alone or with C5a did not damage the cells. Cross-linking of HLA appears to play a role in the ability of antibody to activate exocytosis, because the W6/32 monovalent Fab fragment did not activate VWF release, but the bivalent Fab'2 was effective in triggering exocytosis. To explore the in vivo effects of antibody upon graft injury, we infused W6/32 Fab'2 antibody to human HLA into severe combined immunodeficient/beige mice that had been transplanted with human skin grafts. Antibody to HLA activated exocytosis and inflammation in human skin grafts. Our data show that antibody to host antigens can activate human endothelial cell exocytosis and leukocyte trafficking. By triggering vascular inflammation, antibody activation of exocytosis may play a role in transplant rejection.
Collapse
Affiliation(s)
- Munekazu Yamakuchi
- *Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Nancy C. Kirkiles-Smith
- Departments of Pathology
- Interdepartmental Program in Vascular Biology and Transplantation, Yale University, New Haven, CT 06536-0812
| | - Marcella Ferlito
- *Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | | | - Clare Bao
- *Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | | | | | | | - Jordan S. Pober
- Departments of Pathology
- Dermatology, and
- Immunobiology, and
- Interdepartmental Program in Vascular Biology and Transplantation, Yale University, New Haven, CT 06536-0812
| | - Charles J. Lowenstein
- Departments of Pathology and
- *Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| |
Collapse
|
36
|
de Nadaï P, Chenivesse C, Gilet J, Porte H, Vorng H, Chang Y, Walls AF, Wallaert B, Tonnel AB, Tsicopoulos A, Zerwes HG. CCR5 usage by CCL5 induces a selective leukocyte recruitment in human skin xenografts in vivo. J Invest Dermatol 2006; 126:2057-64. [PMID: 16778803 DOI: 10.1038/sj.jid.5700369] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
CCR5 is one of the major inflammatory chemokine receptors with potential therapeutical applications in humans. However, the redundancy of chemokines and their receptors, and the species specificity of chemokine receptor antagonists pose challenges to understanding of the role they play in pharmacological situations. To address this question, we used a humanized severe combined immunodeficient mouse model grafted with human skin and autologous leukocytes, and evaluated the effect of a blocking antibody against human CCR5, on CCL5-induced cutaneous leukocyte recruitment in vivo. At baseline, CCL5 induced a significant recruitment of T cells mainly of the memory phenotype, of monocytes/macrophages, eosinophils, and IFN-gamma(+) but not IL-4(+) and IL-5(+) cells. In vivo, anti-CCR5 antibody was able to almost completely inhibit the recruitment of monocytes/macrophages and T-helper (Th)1-type cells to inhibit partially the attraction of memory T cells, but had no effect on eosinophil infiltration, although all these cell types express other CCL5 binding chemokine receptors than CCR5. These results indicate that the in vivo environment regulates target cell specificity of CCL5 leading to differential cell recruitment, suggesting that antagonizing CCR5 receptor may be of therapeutic value in diseases such as acquired immuno deficiency syndrome, where CCL5/CCR5, monocytes, and Th1-type cells play a predominant role.
Collapse
|
37
|
Igney FH, Asadullah K, Zollner TM. Humanised mouse models in drug discovery for skin inflammation. Expert Opin Drug Discov 2006; 1:53-68. [DOI: 10.1517/17460441.1.1.53] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
38
|
Yi T, Cuchara L, Wang Y, Koh KP, Ranjbaran H, Tellides G, Pober JS, Lorber MI. Human allograft arterial injury is ameliorated by sirolimus and cyclosporine and correlates with suppression of interferon-gamma. Transplantation 2006; 81:559-66. [PMID: 16495804 DOI: 10.1097/01.tp.0000198737.12507.19] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Chronic allograft dysfunction may result from arterial injury, manifest as transplant arteriosclerosis (TA). This represents an important factor limiting long-term outcomes after heart and kidney transplantation; a relationship between acute allograft arterial injury and TA has been suggested. We have used SCID/bg mice bearing transplanted human artery, inoculated with allogeneic human PBMC to study arteriopathy in human vessels. Earlier work demonstrated arteriopathy similar to that observed clinically, and identified interferon-gamma as a mediator of the process. This study evaluated whether sirolimus (SRL), with cyclosporine A (CsA) or alone, affects TA, and examined possible mechanisms of action. METHODS CB17/SCID/bg mice were transplanted with human arteries replacing the abdominal aorta; reconstituted with allogeneic human PBMC. Controls received vehicle alone for comparison with mice given CsA (5 mg/kg/d), SRL (0.1 or 0.5 mg/kg/d), or CsA (5 mg/kg/d) plus SRL (0.1 mg/kg/d). Transplant arteries were examined 28 days later by histology and immunohistochemistry; circulating human interferon-gamma was evaluated by ELISA, and intragraft interferon-gamma mRNA by qRT-PCR. RESULTS The characteristic TA was modestly reduced by CsA or low-dose SRL, but eliminated by combination CsA plus SRL or higher dose SRL alone. Circulating interferon-gamma was reduced by CsA, but inhibition was dramatic with SRL alone or combined with CsA. Intragraft interferon-gamma and HLA-DR expression were moderately reduced by CsA or SRL, and eliminated with combined CsA plus SRL. CONCLUSIONS SRL plus CsA prevented allograft arteriopathy, correlating with suppression of intragraft interferon-gamma, suggesting that SRL effects may result from anti-inflammatory consequences from inhibiting interferon-gamma.
Collapse
Affiliation(s)
- Tai Yi
- Department of Surgery, Section of Organ Transplantation and Immunology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Shiao SL, McNiff JM, Pober JS. Memory T Cells and Their Costimulators in Human Allograft Injury. THE JOURNAL OF IMMUNOLOGY 2005; 175:4886-96. [PMID: 16210590 DOI: 10.4049/jimmunol.175.8.4886] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Both CD4(+) and CD8(+) human memory but not naive T cells respond to allogeneic human dermal microvascular endothelial cells (HDMEC) in vitro by secreting cytokines and by proliferating. Several recently identified costimulators, namely, 4-1BB ligand, ICOS ligand, and OX40 ligand, are up-regulated on cultured HDMEC in response to TNF or coculture with allogeneic T cells. Blockade of these costimulators each partially reduces IFN-gamma and IL-2 secretion and proliferation of previously resting memory T cells. The effects of these costimulators are overlapping but not identical. Memory but not naive T cells are the principal effectors of microvascular injury in human skin allografts following adoptive transfer into immunodeficient mice. Furthermore, blocking 4-1BB ligand, ICOS ligand, or OX40 ligand in this model reduces human skin allograft injury and T cell effector molecule expression. These data demonstrate that human memory T cells respond to microvascular endothelial cells and can injure allografts in vivo without priming. Furthermore, several recently described costimulators contribute to these processes.
Collapse
Affiliation(s)
- Stephen L Shiao
- Section of Immunobiology, Boyer Center for Molecular Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | |
Collapse
|
40
|
Thomsen M, Yacoub-Youssef H, Marcheix B. Reconstitution of a human immune system in immunodeficient mice: models of human alloreaction in vivo. ACTA ACUST UNITED AC 2005; 66:73-82. [PMID: 16029426 DOI: 10.1111/j.1399-0039.2005.00409.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Rodents have been widely used for studies in transplantation immunology because of their short reproduction period and the relative ease of generating inbred mutant or transgenic strains. However, although many biological mechanisms are similar between rodents and humans, several features clearly distinguish the immune system in these species. Consequently, it is rarely possible to extrapolate observations from rodent models directly into clinical practice. In vitro studies with human cells are useful for elucidation of basic mechanisms, but in order to study complex biological phenomena, in vivo studies are indispensable. In later years, a number of interesting models have been described where immunodeficient mice have been reconstituted with human cells, so-called humanized mice, in order to study human immune responses in vivo. This has opened a new field of experimental immunology that has been applied to areas such as cancer, autoimmunity, allergy, infections, and transplantation biology. In this review, we shall concentrate on the use of severe combined immunodeficient mice reconstituted with human immune or stem cells for studies of human alloreaction in vivo.
Collapse
Affiliation(s)
- M Thomsen
- INSERM U466, CHU Rangueil, Toulouse, France.
| | | | | |
Collapse
|
41
|
Choi J, Walker J, Boichuk S, Kirkiles-Smith N, Torpey N, Pober JS, Alexander L. Human endothelial cells enhance human immunodeficiency virus type 1 replication in CD4+ T cells in a Nef-dependent manner in vitro and in vivo. J Virol 2005; 79:264-76. [PMID: 15596822 PMCID: PMC538695 DOI: 10.1128/jvi.79.1.264-276.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infected CD4+ T cells are the primary sites of human immunodeficiency virus type 1 (HIV-1) replication in vivo. However, signals from professional antigen-presenting cells (APCs), such as dendritic cells and macrophages, greatly enhance HIV-1 replication in T cells. Here, we report that in cocultures, vascular endothelial cells (ECs), which in humans can also serve as APCs, can enhance HIV-1 production of both CCR5- and CXCR4-utilizing strains approximately 50,000-fold. The observed HIV-1 replication enhancement conferred by ECs occurred only in memory CD4+ T cells, required expression of major histocompatibility complex class II (MHC-II) molecules by the ECs, and could not be conferred by fixed ECs, all of which are consistent with a requirement for EC-mediated T-cell activation via T-cell receptor (TCR) signaling. Deletion of nef (Nef-) decreased HIV-1 production by approximately 100-fold in T cells cocultured with ECs but had no effect on virus production in T cells cocultured with professional APCs or fibroblasts induced to express MHC-II. Human ECs do not express B7 costimulators, but Nef- replication in CD4(+)-T-cell and EC cocultures could not be rescued by anti-CD28 antibody. ECs act in trans to enhance wild-type but not Nef- replication and facilitate enhanced wild-type replication in naive T cells when added to T-cell or B-lymphoblastoid cell cocultures, suggesting that ECs also provide a TCR-independent signal to infected T cells. Consistent with these in vitro observations, wild-type HIV-1 replicated 30- to 50-fold more than Nef- in human T cells infiltrating allogeneic human skin grafts on human huPBL-SCID/bg mice, an in vivo model of T-cell activation by ECs. Our studies suggest that ECs, which line the entire cardiovascular system and are, per force, in frequent contact with memory CD4+ T cells, provide signals to HIV-1-infected CD4+ T cells to greatly enhance HIV-1 production in a Nef-dependent manner, a mechanism that could contribute to the development of AIDS.
Collapse
Affiliation(s)
- Jaehyuk Choi
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Enis DR, Shepherd BR, Wang Y, Qasim A, Shanahan CM, Weissberg PL, Kashgarian M, Pober JS, Schechner JS. Induction, differentiation, and remodeling of blood vessels after transplantation of Bcl-2-transduced endothelial cells. Proc Natl Acad Sci U S A 2004; 102:425-30. [PMID: 15625106 PMCID: PMC544288 DOI: 10.1073/pnas.0408357102] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Implants of collagen-fibronectin gels containing Bcl-2-transduced human umbilical vein endothelial cells (Bcl-2-HUVECs) induce the formation of human endothelial cell (EC)/murine vascular smooth muscle cell (VSMC) chimeric vessels in immunodeficient mice. Microfil casting of the vasculature 60 d after implantation reveals highly branched microvascular networks within the implants that connect with and induce remodeling of conduit vessels arising from the abdominal wall circulation. Approximately 85% of vessels within the implants are lined by Bcl-2-positive human ECs expressing VEGFR1, VEGFR2, and Tie-2, but not integrin alpha(v)beta(3). The human ECs are seated on a well formed human laminin/collagen IV-positive basement membrane, and are surrounded by mouse VSMCs expressing SM-alpha actin, SM myosin, SM22alpha, and calponin, all markers of contractile function. Transmission electron microscopy identified well formed EC-EC junctions, chimeric arterioles with concentric layers of contractile VSMC, chimeric capillaries surrounded by pericytes, and chimeric venules. Bcl-2-HUVEC-lined vessels retain 70-kDa FITC-dextran, but not 3-kDa dextran; local histamine rapidly induces leak of 70-kDa FITC-dextran or India ink. As in skin, TNF induces E-selectin and vascular cell adhesion molecule 1 only on venular ECs, whereas intercellular adhesion molecule-1 is up-regulated on all human ECs. Bcl-2-HUVEC implants are able to engraft within and increase perfusion of ischemic mouse gastrocnemius muscle after femoral artery ligation. These studies show that cultured Bcl-2-HUVECs can differentiate into arterial, venular, and capillary-like ECs when implanted in vivo, and induce arteriogenic remodeling of the local mouse vessels. Our results support the utility of differentiated EC transplantation to treat tissue ischemia.
Collapse
Affiliation(s)
- David R Enis
- Interdepartmental Program in Vascular Biology and Transplantation, Boyer Center for Molecular Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ullrich S, Schumacher U, Ai M, Tiemann B, Gay S, Schechner JS, Pober JS, Gross WL, Csernok E. Transfer of Human Leukocytes into Double-Knockout Pfp−/−Rag2−/− Mice Grafted with Human Skin: Increased Accumulation of Neutrophils in Human Dermal Microvessels. Transplantation 2004; 78:1557-9. [PMID: 15599322 DOI: 10.1097/01.tp.0000144326.75485.b9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Severe combined immunodeficient mice reconstituted with human leukocytes have been useful to model parts of the human immune system, including some of its diseases (e.g., AIDS). Because no human polymorphonuclear leukocytes (huPMN) develop in these xenograft models, diseases such as several forms of vasculitis cannot be modeled using this approach. To provide such a model for vasculitis, human skin patches were grafted onto double-knockout Pfp(-/-)Rag2(-/-) mice, which not only lack functional T and B cells but which are also devoid of natural killer cells. After intravenous injection, a high proportion of huPMNs survived within the circulation and accumulated in the human blood vessels. The accumulation increased considerably after the endothelium of the skin patches had been stimulated by tumor necrosis factor-alpha. Alpha mild perivascular neutrophilic infiltration and vascular necrosis was observed in the microvessels of the skin patches. Thus, a xenograft model of vasculitis with predominant huPMNs infiltration has been established for the first time.
Collapse
Affiliation(s)
- Sebastian Ullrich
- Department of Rheumatology, University Hospital of Schleswig-Holstein, Campus Lübeck and Rheumaklinik Bad Bramstedt, Ratzeburger Alle 160, 23538 Lübeck, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Banuelos SJ, Shultz LD, Greiner DL, Burzenski LM, Gott B, Lyons BL, Rossini AA, Appel MC. Rejection of human islets and human HLA-A2.1 transgenic mouse islets by alloreactive human lymphocytes in immunodeficient NOD-scid and NOD-Rag1nullPrf1null mice. Clin Immunol 2004; 112:273-83. [PMID: 15308121 DOI: 10.1016/j.clim.2004.04.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2004] [Accepted: 04/19/2004] [Indexed: 11/23/2022]
Abstract
Immunodeficient NOD mice engrafted with human peripheral blood mononuclear cells (PBMCs) were used in two models of human islet allograft rejection. Model one: human PBMCs were engrafted into chemically diabetic NOD-scid mice bearing established subrenal human islet allografts. Inflammation and often complete islet allograft rejection were observed. Model 2 incorporated three key advances. First, we developed a new immunodeficient recipient, NOD-RagI(null)Prf1(null) mice. Second, graft-lymphocyte interactions were optimized by intrasplenic co-transplantation of islets and human PBMC. Third, NOD-scid islets expressing human HLA-A2.1 were used as allograft targets. Diabetic NOD-RagI(null)Prf1(null) recipients of HLA-A2.1 transgenic mouse islets, alone or co-engrafted with HLA-A2-positive human PBMC, exhibited durable graft survival and euglycemia. Contrastingly, co-transplantation with HLA-A2-negative human PBMC led to islet graft rejection without evidence of graft-vs.-host disease (GVHD). We propose that diabetic NOD-RagI(null)Prf1(null) mice co-engrafted with HLA-A2 mouse transgenic islets and allogeneic human PBMC provide an effective in vivo model of human islet allograft rejection.
Collapse
Affiliation(s)
- Scott J Banuelos
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Snanoudj R, Rouleau M, Bidère N, Carmona S, Baron C, Latinne D, Bazin H, Charpentier B, Senik A. A Role for CD2 Antibodies (BTI-322 and its Humanized Form) in the in vivo Elimination of Human T Lymphocytes Infiltrating an Allogeneic Human Skin Graft in SCID Mice: An Fcγ Receptor-Related Mechanism Involving Co-Injected Human NK Cells. Transplantation 2004; 78:50-8. [PMID: 15257038 DOI: 10.1097/01.tp.0000128235.04297.43] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Pilot clinical studies have shown that the rat anti-human-CD2 monoclonal antibody, LoCD2a/BTI-322, can efficiently prevent and treat acute kidney rejection. However, the in vivo mechanism by which it prevents allograft rejection has not been studied. BTI-322 and its humanized form have been shown to mediate in vitro antibody-dependent cell-mediated cytotoxicity (ADCC) against CD2 cells through the activation of monocytes or natural killer (NK) cells. METHODS Human fetal skin samples were grafted into severe combined immunodeficient/nonobese diabetic mice. Five weeks later (day 0), the mice were injected with human allogeneic peripheral blood lymphocytes (PBL). Either on day 0 or on day 14, mice were treated with BTI-322, hu-BTI-322, or their F(ab')2 fragments. Peripheral blood mononuclear cells (PBMC) thoroughly devoid of NK cells were also assayed. RESULTS After injection of PBL, the human skins became heavily infiltrated with activated human T lymphocytes, resulting in dermal microvascular injuries indicative of graft rejection. Early treatment with BTI-322 and hu-BTI-322 prevented all these events. These CD2 antibodies rapidly eliminated human T lymphocytes that had already infiltrated the grafts, with no evidence of recirculation toward the spleen. Their F(ab')2 fragments were, in contrast, ineffective. Elimination of NK cells from injected PBMC prevented the curative effect exerted by whole CD2 antibodies. It also abrogated their cytotoxicity potential against CD2 cells in ADCC assays. CONCLUSION F(ab')2 fragments of the CD2 antibodies could not prevent allograft rejection, whereas whole immunoglobulin G could, and human NK cells were required for the curative effect exerted by these antibodies. The results are consistent with an FcgammaR-dependent ADCC mechanism mediated in vivo by human NK cells.
Collapse
|
46
|
Sadeghi MM, Schechner JS, Krassilnikova S, Gharaei AA, Zhang J, Kirkiles-Smith N, Sinusas AJ, Zaret BL, Bender JR. Vascular cell adhesion molecule-1-targeted detection of endothelial activation in human microvasculature. Transplant Proc 2004; 36:1585-91. [PMID: 15251390 DOI: 10.1016/j.transproceed.2004.05.060] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The hallmark of endothelial activation, an early and critical step in many alloimmune and inflammatory responses, is the transcriptional induction and expression of endothelial adhesion molecules (eg, vascular cell adhesion molecule-1 [VCAM-1]). We assessed the feasibility of VCAM-1-targeted in vivo detection of endothelial activation using I-125-labeled-F(ab')2 fragments of E1/6, a monoclonal antibody against human but not murine VCAM-1. The Kd and Bmax, determined by saturation binding in tumor necrosis factor (TNF)-activated human endothelial cells (ECs), were 3.2 +/- 0.6 nmol/L and 5600 +/- 300 binding sites per EC, respectively. Biodistribution and in vivo binding characteristics of I-125-E1/6 F(ab')2 were assessed in a novel chimeric human/mouse model, in which human skin (as a source of human microvasculature) is grafted onto SCID/beige mice. I-125-E1/6 F(ab')2 localized to TNF-activated human skin grafts as detected by autoradiography and gamma well-counting. Relative uptakes (uptake in human skin graft/uptake in the surrounding mouse skin) were, respectively, 2.6 +/- 0.8 (n = 14) and 1.6 +/- 0.3 (n = 12) for E1/6 and MOPC-21, an isotype-matched control antibody (P < .01). The preferential uptake in human skin graft was not due to differences in tissue vascularity assessed by Tc-99m-labeled murine red blood cells. In conclusion, the chimeric human/mouse model is a novel experimental tool for in vivo evaluation of human endothelial cell-specific radiopharmaceuticals. Although I-125-E1/6 F(ab')2 localized to human skin grafts, the limited number of VCAM-1 molecules/endothelial cell adversely affects its suitability as a target for in vivo imaging of endothelial activation.
Collapse
Affiliation(s)
- M M Sadeghi
- Raymond and Beverly Sackler Cardiovascular Molecular Imaging Laboratory, Department of Dermatology, Boyer Center for Molecular Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kirkiles-Smith NC, Mahboubi K, Plescia J, McNiff JM, Karras J, Schechner JS, Altieri DC, Pober JS. IL-11 protects human microvascular endothelium from alloinjury in vivo by induction of survivin expression. THE JOURNAL OF IMMUNOLOGY 2004; 172:1391-6. [PMID: 14734714 DOI: 10.4049/jimmunol.172.3.1391] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-11 can reduce tissue injury in animal models of inflammation but the mechanism(s) is unknown. When C.B-17 SCID/beige mice bearing human skin grafts are injected i.p. with human PBMC allogeneic to the donor skin, infiltrating T cells destroy human microvessels by day 21. Intradermal injection of human IL-11 (500 ng/day) delays the time course of graft microvessel loss without reducing the extent of T cell infiltration. Protective actions of IL-11 are most pronounced on day 15. IL-11 has no effect on T cell activation marker, effector molecule, cytokine expression, or endothelial ICAM-1 expression. IL-11 up-regulates the expression of survivin, a cytoprotective protein, in graft keratinocytes and endothelial cells. Topical application of survivin antisense oligonucleotide down-regulates survivin expression in both cell types and largely abrogates the protective effect of IL-11. We conclude that in this human transplant model, IL-11 exerts a cytoprotective rather than anti-inflammatory or immunomodulatory effect mediated through induction of survivin.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Topical
- Adult
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Cytoprotection/genetics
- Cytoprotection/immunology
- Down-Regulation/genetics
- Down-Regulation/immunology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Humans
- Inhibitor of Apoptosis Proteins
- Injections, Intradermal
- Interleukin-11/administration & dosage
- Interleukin-11/antagonists & inhibitors
- Interleukin-11/therapeutic use
- Leukocytes, Mononuclear/transplantation
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, SCID
- Microcirculation/immunology
- Microcirculation/metabolism
- Microcirculation/pathology
- Microtubule-Associated Proteins/antagonists & inhibitors
- Microtubule-Associated Proteins/biosynthesis
- Microtubule-Associated Proteins/genetics
- Microtubule-Associated Proteins/physiology
- Neoplasm Proteins
- Ointments
- Oligonucleotides, Antisense/administration & dosage
- Skin/blood supply
- Skin/immunology
- Skin/metabolism
- Skin/pathology
- Skin Transplantation/adverse effects
- Skin Transplantation/immunology
- Skin Transplantation/pathology
- Survivin
- Transplantation, Homologous/adverse effects
- Transplantation, Homologous/immunology
- Transplantation, Homologous/pathology
Collapse
Affiliation(s)
- Nancy C Kirkiles-Smith
- Interdepartmental Program in Vascular Biology and Transplantation, Boyer Center for Molecular Medicine and Department of Pathology, Yale University School of Medicine, New Haven CT 06510, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Reinders MEJ, Sho M, Izawa A, Wang P, Mukhopadhyay D, Koss KE, Geehan CS, Luster AD, Sayegh MH, Briscoe DM. Proinflammatory functions of vascular endothelial growth factor in alloimmunity. J Clin Invest 2004; 112:1655-65. [PMID: 14660742 PMCID: PMC281640 DOI: 10.1172/jci17712] [Citation(s) in RCA: 189] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF), an established angiogenesis factor, is expressed in allografts undergoing rejection, but its function in the rejection process has not been defined. Here, we initially determined that VEGF is functional in the trafficking of human T cells into skin allografts in vivo in the humanized SCID mouse. In vitro, we found that VEGF enhanced endothelial cell expression of the chemokines monocyte chemoattractant protein 1 and IL-8, and in combination with IFN-gamma synergistically induced endothelial cell production of the potent T cell chemoattractant IFN-inducible protein-10 (IP-10). Treatment of BALB/c (H-2d) recipients of fully MHC-mismatched C57BL/6 (H-2b) donor hearts with anti-VEGF markedly inhibited T cell infiltration of allografts and acute rejection. Anti-VEGF failed to inhibit T cell activation responses in vivo, but inhibited intragraft expression of several endothelial cell adhesion molecules and chemokines, including IP-10. In addition, whereas VEGF expression was increased, neovascularization was not associated with acute rejection, and treatment of allograft recipients with the angiogenesis inhibitor endostatin failed to inhibit leukocyte infiltration of the grafts. Thus, VEGF appears to be functional in acute allograft rejection via its effects on leukocyte trafficking. Together, these observations provide mechanistic insight into the proinflammatory function of VEGF in immunity.
Collapse
Affiliation(s)
- Marlies E J Reinders
- Division of Nephrology, Department of Medicine, Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Turgeon NA, Banuelos SJ, Shultz LD, Lyons BL, Iwakoshi N, Greiner DL, Mordes JP, Rossini AA, Appel MC. Alloimmune injury and rejection of human skin grafts on human peripheral blood lymphocyte-reconstituted non-obese diabetic severe combined immunodeficient beta2-microglobulin-null mice. Exp Biol Med (Maywood) 2003; 228:1096-104. [PMID: 14530522 DOI: 10.1177/153537020322800918] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Small animal models with the capacity to support engraftment of a functional human immune system are needed to facilitate studies of human alloimmunity. In the present investigation, non-obese diabetic (NOD) severe combined immunodeficient (scid) beta2-microglobulin-null (B2mnull) mice engrafted with human peripheral blood lymphocytes (hu-PBL-NOD-scid B2mnull mice) were used as in vivo models for studying human skin allograft rejection. Hu-PBL-NOD-scid B2mnull mice were established by injection of human spleen cells or PBLs and transplanted with full-thickness allogeneic human skin. Human cell engraftment was enhanced by injection of anti-mouse CD122 antibody. The respective contributions of human CD4+ and CD8+ cells in allograft rejection were determined using depleting antibodies. Human skin grafts on unmanipulated NOD-scid B2mnull mice uniformly survived but on chimeric hu-PBL-NOD-scid B2mnull mice exhibited severe immune-mediated injury that often progressed to complete rejection. The alloaggressive hu-PBLs did not require prior in vitro sensitization to elicit targeted effector cell activity. Extensive mononuclear cell infiltration directed towards human-origin endothelium was associated with thrombosis and fibrin necrosis. No evidence of graft-versus-host disease was detected. Either CD4+ or CD8+ T cells may mediate injury and alloimmune rejection of human skin grafts on hu-PBL-NOD-scid B2mnull mice. It is proposed that Hu-PBL-NOD-scid B2mnull mice engrafted with human skin will provide a useful model for analysis of interventions designed to modulate human allograft rejection.
Collapse
Affiliation(s)
- Nicole A Turgeon
- University of Massachusetts Medical School, Worcester, Massachussetts 01605, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Li JH, Kirkiles-Smith NC, McNiff JM, Pober JS. TRAIL induces apoptosis and inflammatory gene expression in human endothelial cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:1526-33. [PMID: 12874246 DOI: 10.4049/jimmunol.171.3.1526] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human TRAIL can efficiently kill tumor cells in vitro and kill human tumor xenografts in mice with little effect on normal mouse cells or tissues. The effects of TRAIL on normal human tissues have not been described. In this study, we report that endothelial cells (EC), isolated from human umbilical veins or human dermal microvessels, express death domain-containing TRAIL-R1 and -R2. Incubation with TRAIL for 15 h causes approximately 30% of cultured EC to die, as assessed by propidium iodide uptake. Death is apoptotic, as assessed by Annexin V staining, 4',6'-diamidino-2-phenylindole staining, and DNA fragment ELISA. EC death is increased by cotreatment with cycloheximide but significantly reduced by caspase inhibitors or transduced dominant-negative Fas-associated death domain protein. In surviving cells, TRAIL activates NF-kappaB, induces expression of E-selectin, ICAM-1, and IL-8, and promotes adhesion of leukocytes. Injection of TRAIL into human skin xenografts promotes focal EC injury accompanied by limited neutrophil infiltration. These data suggest that TRAIL is an inducer of tissue injury in humans, an outcome that may influence antitumor therapy with TRAIL.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Apoptosis/physiology
- Apoptosis Regulatory Proteins
- Cell Line
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiology
- Gene Expression Regulation/physiology
- HL-60 Cells
- HeLa Cells
- Humans
- Inflammation Mediators/administration & dosage
- Inflammation Mediators/metabolism
- Inflammation Mediators/pharmacology
- Injections, Intradermal
- Leukocytes, Mononuclear/pathology
- Membrane Glycoproteins/administration & dosage
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, SCID
- Neutrophil Infiltration/physiology
- Receptors, TNF-Related Apoptosis-Inducing Ligand
- Receptors, Tumor Necrosis Factor/biosynthesis
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/pharmacology
- Skin Transplantation/pathology
- TNF-Related Apoptosis-Inducing Ligand
- Transplantation, Heterologous/pathology
- Tumor Necrosis Factor-alpha/administration & dosage
- Tumor Necrosis Factor-alpha/physiology
Collapse
Affiliation(s)
- Jie Hui Li
- Interdepartmental Program in Vascular Biology and Transplantation, Boyer Center for Molecular Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|