1
|
Boiko JR, Hill GR. Chronic Graft-versus-host Disease: Immune Insights, Therapeutic Advances, and Parallels for Solid Organ Transplantation. Transplantation 2024:00007890-990000000-00959. [PMID: 39682018 DOI: 10.1097/tp.0000000000005298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Chronic graft-versus-host disease remains a frequent and morbid outcome of allogeneic hematopoietic cell transplantation, in which the donor-derived immune system attacks healthy recipient tissue. Preceding tissue damage mediated by chemoradiotherapy and alloreactive T cells compromise central and peripheral tolerance mechanisms, leading to aberrant donor T cell and germinal center B cell differentiation, culminating in pathogenic macrophage infiltration and differentiation in a target tissue, with ensuant fibrosis. This process results in a heterogeneous clinical syndrome with significant morbidity and mortality, frequently requiring prolonged therapy. In this review, we discuss the processes that interrupt immune tolerance, the subsequent clinical manifestations, and new Food and Drug Administration-approved therapeutic approaches that have been born from a greater understanding of disease pathogenesis in preclinical systems, linking to parallel processes following solid organ transplantation.
Collapse
Affiliation(s)
- Julie R Boiko
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Geoffrey R Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
2
|
Jeong SY, Park D, Park T, Han JS, Lee J, Choi CH, Jo M, Lee YB, Kyun ML, Choi M, Park D, Moon KS. Interspecies transcriptome profiles of human T cell activation and liver inflammation in a xenogeneic graft-versus-host disease model. Heliyon 2024; 10:e40559. [PMID: 39687194 PMCID: PMC11648781 DOI: 10.1016/j.heliyon.2024.e40559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Background Xenogeneic transplantation induces acute graft-versus-host disease (aGvHD) and subsequent vital organ damage. Herein, we aimed to examine hepatic damage associated with aGvHD using histopathology and gene expression profiles. Methods A xenografic GvHD model was established by engrafting human peripheral blood mononuclear cells (PBMCs) into immunodeficient NOD-scid IL2Rγnull (NSG) mice after busulfan conditioning. NSG mice were assigned to groups treated with saline (S group) or a combination of busulfan and PBMCs (BP group). Histological lesions and RNA sequencing analysis of gene profiles in the BP group (GvHD model) were compared with those in the P group. Results Predominant T cell subsets (95 %) in the blood of the BP group were identified as cytotoxic CD8+ T cells (56 %) and helper CD4+ T cells (31 %). Symptoms of aGvHD, including hepatocyte necrosis, bile duct hyperplasia, and human T cell infiltration, were observed. Gene expression analysis revealed upregulation of Th1 and Th2 cell differentiation (STAT4, IL4R, and NFACT1), T cell receptor signaling pathway (CD226 and GBP1), IL-1 pathway (CCL3, NAIP, and IRAK4), cell cycle (CDCA5, CDCA8, MCM5, KNL1, BUB1B, FBXO5, and CENPE) in human cells. In mouse cells, Il1a, Ifngr, Tnfrsf, and Il6ra genes (cytokines or their receptors) and Icam, Vcam, and Endra genes (adhesion molecules) were upregulated, whereas genes related to chromosome condensation (H2ac and H2bc) and fatty acid/steroid metabolism (Fasn, Rdh, and Scd) were downregulated. Interspecies gene network analysis revealed that activated human T cells are associated with liver damage through inflammatory and metabolic pathways, accompanied by increased mouse cell adhesion molecules and cytokines. Conclusion Our findings offer valuable insights into the pathophysiology and biomarkers of aGvHD and may contribute to the development of novel therapeutics.
Collapse
Affiliation(s)
- Seo Yule Jeong
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Duhyeon Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Tamina Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Ji-Seok Han
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Jungyun Lee
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Chang Hoon Choi
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Minseong Jo
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Yu Bin Lee
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Mi-lang Kyun
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Myeongjin Choi
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Daeui Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Kyoung-Sik Moon
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| |
Collapse
|
3
|
Ménoret S, Renart-Depontieu F, Martin G, Thiam K, Anegon I. Efficient generation of human immune system rats using human CD34 + cells. Stem Cell Reports 2024; 19:1255-1263. [PMID: 39151431 PMCID: PMC11411320 DOI: 10.1016/j.stemcr.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 07/20/2024] [Accepted: 07/21/2024] [Indexed: 08/19/2024] Open
Abstract
Human immune system (HIS) mice generated using human CD34+ hematopoietic stem cells serve as a pivotal model for the in vivo evaluation of immunotherapies for humans. Yet, HIS mice possess certain limitations. Rats, due to their size and comprehensive immune system, hold promise for translational experiments. Here, we describe an efficacious method for long-term immune humanization, through intrahepatic injection of hCD34+ cells in newborn immunodeficient rats expressing human SIRPα. In contrast to HIS mice and similar to humans, HIS rats showed in blood a predominance of T cells, followed by B cells. Immune humanization was also high in central and secondary lymphoid organs. HIS rats treated with the anti-human CD3 antibody were depleted of human T cells, and human cytokines were detected in sera. We describe for the first time a method to efficiently generate HIS rats. HIS rats have the potential to be a useful model for translational immunology.
Collapse
Affiliation(s)
- Séverine Ménoret
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, F-44000 Nantes, France; INSERM, Centre de Recherche en Transplantation et Immunologie UMR1064, Nantes Université, Nantes, France.
| | | | | | | | - Ignacio Anegon
- INSERM, Centre de Recherche en Transplantation et Immunologie UMR1064, Nantes Université, Nantes, France.
| |
Collapse
|
4
|
Celhar T, Li X, Zhao Y, Tay HC, Lee A, Liew HH, Shepherdson EK, Rajarethinam R, Fan Y, Mak A, Chan JKY, Singhal A, Takahashi T. Fetal liver CD34 + contain human immune and endothelial progenitors and mediate solid tumor rejection in NOG mice. Stem Cell Res Ther 2024; 15:164. [PMID: 38853275 PMCID: PMC11163708 DOI: 10.1186/s13287-024-03756-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/07/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Transplantation of CD34+ hematopoietic stem and progenitor cells (HSPC) into immunodeficient mice is an established method to generate humanized mice harbouring a human immune system. Different sources and methods for CD34+ isolation have been employed by various research groups, resulting in customized models that are difficult to compare. A more detailed characterization of CD34+ isolates is needed for a better understanding of engraftable hematopoietic and potentially non-hematopoietic cells. Here we have performed a direct comparison of CD34+ isolated from cord blood (CB-CD34+) or fetal liver (FL-CD34+ and FL-CD34+CD14-) and their engraftment into immunocompromised NOD/Shi-scid Il2rgnull (NOG) mice. METHODS NOG mice were transplanted with either CB-CD34+, FL-CD34+ or FL-CD34+CD14- to generate CB-NOG, FL-NOG and FL-CD14--NOG, respectively. After 15-20 weeks, the mice were sacrificed and human immune cell reconstitution was assessed in blood and several organs. Liver sections were pathologically assessed upon Haematoxylin and Eosin staining. To assess the capability of allogenic tumor rejection in CB- vs. FL-reconstituted mice, animals were subcutaneously engrafted with an HLA-mismatched melanoma cell line. Tumor growth was assessed by calliper measurements and a Luminex-based assay was used to compare the cytokine/chemokine profiles. RESULTS We show that CB-CD34+ are a uniform population of HSPC that reconstitute NOG mice more rapidly than FL-CD34+ due to faster B cell development. However, upon long-term engraftment, FL-NOG display increased numbers of neutrophils, dendritic cells and macrophages in multiple tissues. In addition to HSPC, FL-CD34+ isolates contain non-hematopoietic CD14+ endothelial cells that enhance the engraftment of the human immune system in FL-NOG mice. We demonstrate that these CD14+CD34+ cells are capable of reconstituting Factor VIII-producing liver sinusoidal endothelial cells (LSEC) in FL-NOG. However, CD14+CD34+ also contribute to hepatic sinusoidal dilatation and immune cell infiltration, which may culminate in a graft-versus-host disease (GVHD) pathology upon long-term engraftment. Finally, using an HLA-A mismatched CDX melanoma model, we show that FL-NOG, but not CB-NOG, can mount a graft-versus-tumor (GVT) response resulting in tumor rejection. CONCLUSION Our results highlight important phenotypical and functional differences between CB- and FL-NOG and reveal FL-NOG as a potential model to study hepatic sinusoidal dilatation and mechanisms of GVT.
Collapse
Affiliation(s)
- Teja Celhar
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore.
- Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan.
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Republic of Singapore.
| | - Xinyi Li
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore
- Interdisciplinary Life Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Yunqian Zhao
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore
| | - Hui Chien Tay
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore
| | - Andrea Lee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Republic of Singapore
| | - Hui Hua Liew
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Republic of Singapore
| | - Edwin Kunxiang Shepherdson
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Republic of Singapore
| | - Ravisankar Rajarethinam
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Yiping Fan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Republic of Singapore
- Obstetrics and Gynaecology Academic Clinical Programme, Duke-NUS Medical School, Singapore, 169857, Republic of Singapore
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, Singapore, 117597, Republic of Singapore
| | - Anselm Mak
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Rheumatology, University Medicine Cluster, National University Health System, Singapore, Republic of Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Republic of Singapore
- Obstetrics and Gynaecology Academic Clinical Programme, Duke-NUS Medical School, Singapore, 169857, Republic of Singapore
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, Singapore, 117597, Republic of Singapore
| | - Amit Singhal
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #04-06, Singapore, 138648, Republic of Singapore
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos #05-13, Singapore, 138648, Republic of Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Republic of Singapore
| | - Takeshi Takahashi
- Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan
| |
Collapse
|
5
|
Peng D, Bai W, Zhang C, Chang X, Ma P, Wang X, Sun S, Zhan L. X-ray irradiation effectively inactivated lymphocytes in transfusion in vivo monitored by the bioluminescence transfusion-associated graft-versus-host disease model. Vox Sang 2024; 119:181-192. [PMID: 38226529 DOI: 10.1111/vox.13559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/15/2023] [Accepted: 10/05/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND AND OBJECTIVES Transfusion of cold-stored whole blood is the preferred resuscitation method for trauma patients but may cause transfusion-associated graft-versus-host disease (TA-GVHD). Standard clinical practice to prevent this is to irradiate blood components with gamma-rays. X-ray irradiations are also a safe and effective alternative to gamma-ray irradiation. We established a visual mouse model of TA-GVHD to compare the viability and function of lymphocytes exposed to gamma- and x-ray irradiation. MATERIALS AND METHODS A haploidentical transplantation mouse model was established to simulate TA-GVHD with Balb/c mice as donors and hybrid F1 CB6 mice (Balb/c × C57) as recipients. Spleen cells from Tg-Fluc+ Balb/c mice were isolated and irradiated with gamma-rays and x-rays. Lymphocyte activation, apoptosis and proliferation post phorbol 1 2-myristate 1 3-acetate (PMA) stimulation were evaluated. After transfusion, we monitored Fluc+ lymphocytes daily by bioluminescence imaging. Recipients were euthanized on day 21, and tissues were examined pathologically and for inflammatory cytokines. RESULTS The viability of gamma- or x-ray irradiated lymphocytes decreased significantly with slight changes in proliferation in vivo after transfusion. Compared with the non-irradiated group, both the gamma- and x-ray irradiated groups showed significantly decreased clinical scoring and inflammatory cytokine levels. The fluorescence intensity of the body and target organs was reduced after irradiation. CONCLUSION No recipients acquired TA-GVHD after lymphocyte transfusion subjected to gamma- or x-rays, showing that x-rays inactivate as well as gamma rays and are suitable for irradiating whole blood.
Collapse
Affiliation(s)
- Dongxin Peng
- Institute of Health Service and Transfusion Medicine, Beijing, People's Republic of China
| | - Wenyuan Bai
- Institute of Health Service and Transfusion Medicine, Beijing, People's Republic of China
| | - Can Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, People's Republic of China
- School of Biomedical Engineering, Anhui Medical University, Hefei, People's Republic of China
| | - Xindai Chang
- Institute of Health Service and Transfusion Medicine, Beijing, People's Republic of China
- School of Materials Science and Engineering, Yanshan University, Qinhuangdao, People's Republic of China
| | - Ping Ma
- Institute of Health Service and Transfusion Medicine, Beijing, People's Republic of China
| | - Xiaohui Wang
- Institute of Health Service and Transfusion Medicine, Beijing, People's Republic of China
| | - Sujing Sun
- Institute of Health Service and Transfusion Medicine, Beijing, People's Republic of China
| | - Linsheng Zhan
- Institute of Health Service and Transfusion Medicine, Beijing, People's Republic of China
| |
Collapse
|
6
|
Bonatti M, Pitozzi V, Caruso P, Pontis S, Pittelli MG, Frati C, Mangiaracina C, Lagrasta CAM, Quaini F, Cantarella S, Ottonello S, Villetti G, Civelli M, Montanini B, Trevisani M. Time-course transcriptome analysis of a double challenge bleomycin-induced lung fibrosis rat model uncovers ECM homoeostasis-related translationally relevant genes. BMJ Open Respir Res 2023; 10:e001476. [PMID: 37730279 PMCID: PMC10510891 DOI: 10.1136/bmjresp-2022-001476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 08/30/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is an irreversible disorder with a poor prognosis. The incomplete understanding of IPF pathogenesis and the lack of accurate animal models is limiting the development of effective treatments. Thus, the selection of clinically relevant animal models endowed with similarities with the human disease in terms of lung anatomy, cell biology, pathways involved and genetics is essential. The bleomycin (BLM) intratracheal murine model is the most commonly used preclinical assay to evaluate new potential therapies for IPF. Here, we present the findings derived from an integrated histomorphometric and transcriptomic analysis to investigate the development of lung fibrosis in a time-course study in a BLM rat model and to evaluate its translational value in relation to IPF. METHODS Rats were intratracheally injected with a double dose of BLM (days 0-4) and sacrificed at days 7, 14, 21, 28 and 56. Histomorphometric analysis of lung fibrosis was performed on left lung sections. Transcriptome profiling by RNAseq was performed on the right lung lobes and results were compared with nine independent human gene-expression IPF studies. RESULTS The histomorphometric and transcriptomic analyses provided a detailed overview in terms of temporal gene-expression regulation during the establishment and repair of the fibrotic lesions. Moreover, the transcriptomic analysis identified three clusters of differentially coregulated genes whose expression was modulated in a time-dependent manner in response to BLM. One of these clusters, centred on extracellular matrix (ECM)-related process, was significantly correlated with histological parameters and gene sets derived from human IPF studies. CONCLUSIONS The model of lung fibrosis presented in this study lends itself as a valuable tool for preclinical efficacy evaluation of new potential drug candidates. The main finding was the identification of a group of persistently dysregulated genes, mostly related to ECM homoeostasis, which are shared with human IPF.
Collapse
Affiliation(s)
- Martina Bonatti
- Department of Chemistry Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Department of Medicine Solna (MedS) and Center for Molecular Medicine (CMM), Karolinska Institutet, Solna, Sweden
| | - Vanessa Pitozzi
- Corporate Preclinical R&D, Chiesi Farmaceutici SpA, Parma, Italy
| | - Paola Caruso
- Corporate Preclinical R&D, Chiesi Farmaceutici SpA, Parma, Italy
| | - Silvia Pontis
- Corporate Preclinical R&D, Chiesi Farmaceutici SpA, Parma, Italy
| | | | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | | | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Simona Cantarella
- Department of Chemistry Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- DKFZ - German Cancer Research Center, Heidelberg, Germany
| | - Simone Ottonello
- Department of Chemistry Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gino Villetti
- Corporate Preclinical R&D, Chiesi Farmaceutici SpA, Parma, Italy
| | - Maurizio Civelli
- Corporate Preclinical R&D, Chiesi Farmaceutici SpA, Parma, Italy
| | - Barbara Montanini
- Department of Chemistry Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Interdepartmental Research Centre Biopharmanet-Tec, University of Parma, Parma, Italy
| | | |
Collapse
|
7
|
Blümich S, Zdimerova H, Münz C, Kipar A, Pellegrini G. Human CD34 + Hematopoietic Stem Cell-Engrafted NSG Mice: Morphological and Immunophenotypic Features. Vet Pathol 2020; 58:161-180. [PMID: 32901581 DOI: 10.1177/0300985820948822] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunodeficient mice engrafted with human immune cells represent an innovative tool to improve translatability of animal models for the study of human diseases. Immunophenotyping in these mice focuses on engraftment rates and cellular differentiation in blood and secondary lymphoid organs, and is predominantly carried out by FACS (fluorescent activated cell sorting) analysis; information on the morphological aspects of engraftment and the prevalence of histologic lesions is limited. We histologically examined 3- to 6-month-old NSG mice, naïve or engrafted with CD34+ human hemopoietic stem cells (HSC), and employed a quantitative immunohistochemical approach to identify human and murine cell compartments, comparing the results with the FACS data. NSG mice mainly exhibited incidental findings in lungs, kidneys, testes, and adrenal glands. A 6-month-old NSG mouse had a mediastinal lymphoblastic lymphoma. The lymphoid organs of NSG mice lacked typical lymphoid tissue architecture but frequently exhibited small periarteriolar leukocyte clusters in the spleen. Mice engrafted with human HSC frequently showed nephropathy, ovarian atrophy, cataract, and abnormal retinal development, lesions considered secondary to irradiation. In addition, 20% exhibited multisystemic granulomatous inflammatory infiltrates, dominated by human macrophages and T cells, leading to the observed 7% mortality and morbidity. Immunophenotypic data revealed variable repopulation of lymphoid organs with hCD45+ human cells, which did not always parallel the engraftment levels measured via FACS. The study describes the most common pathological features in young NSG mice after human HSC engraftment. As some of these lesions contribute to morbidity, morphological assessment of the engraftment at tissue level might help improve immunophenotypic evaluations of this animal model.
Collapse
Affiliation(s)
- Sandra Blümich
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, 27217University of Zurich, Zurich, Switzerland
| | - Hana Zdimerova
- Viral Immunobiology, Institute of Experimental Immunology, 27217University of Zurich, Zurich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, 27217University of Zurich, Zurich, Switzerland
| | - Anja Kipar
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, 27217University of Zurich, Zurich, Switzerland
| | - Giovanni Pellegrini
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, 27217University of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Fukasaku Y, Goto R, Ganchiku Y, Emoto S, Zaitsu M, Watanabe M, Kawamura N, Fukai M, Shimamura T, Taketomi A. Novel immunological approach to asses donor reactivity of transplant recipients using a humanized mouse model. Hum Immunol 2020; 81:342-353. [PMID: 32345498 DOI: 10.1016/j.humimm.2020.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/04/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022]
Abstract
In organ transplantation, a reproducible and robust immune-monitoring assay has not been established to determine individually tailored immunosuppressants (IS). We applied humanized mice reconstituted with human (hu-) peripheral blood mononuclear cells (PBMCs) obtained from living donor liver transplant recipients to evaluate their immune status. Engraftment of 2.5 × 106 hu-PBMCs from healthy volunteers and recipients in the NSG mice was achieved successfully. The reconstituted lymphocytes consisted mainly of hu-CD3+ lymphocytes with predominant CD45RA-CD62Llo TEM and CCR6-CXCR3+CD4+ Th1 cells in hu-PBMC-NSG mice. Interestingly, T cell allo-reactivity of hu-PBMC-NSG mice was amplified significantly compared with that of freshly isolated PBMCs (p < 0.05). Furthermore, magnified hu-T cell responses to donor antigens (Ag) were observed in 2/10 immunosuppressed recipients with multiple acute rejection (AR) experiences, suggesting that the immunological assay in hu-PBMC-NSG mice revealed hidden risks of allograft rejection by IS. Furthermore, donor Ag-specific hyporesponsiveness was maintained in recipients who had been completely weaned off IS (n = 4), despite homeostatic proliferation of hu-T cells in the hu-PBMC-NSG mice. The immunological assay in humanized mice provides a new tool to assess recipient immunity in the absence of IS and explore the underlying mechanisms to maintaining operational tolerance.
Collapse
Affiliation(s)
- Yasutomo Fukasaku
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan
| | - Ryoichi Goto
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan.
| | - Yoshikazu Ganchiku
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan
| | - Shin Emoto
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan
| | - Masaaki Zaitsu
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan
| | - Masaaki Watanabe
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan; Department of Transplant Surgery, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan
| | - Norio Kawamura
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan; Department of Transplant Surgery, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan
| | - Moto Fukai
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan
| | - Tsuyoshi Shimamura
- Division of Organ Transplantation, Hokkaido University Hospital, Sapporo 060-8648, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo 060-8648, Japan.
| |
Collapse
|
9
|
Chun S, Phan MTT, Hong S, Yang J, Yoon Y, Han S, Kang J, Yazer MH, Kim J, Cho D. Double-filtered leukoreduction as a method for risk reduction of transfusion-associated graft-versus-host disease. PLoS One 2020; 15:e0229724. [PMID: 32214402 PMCID: PMC7098637 DOI: 10.1371/journal.pone.0229724] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/13/2020] [Indexed: 11/20/2022] Open
Abstract
Background Transfusion-associated graft-versus-host disease (TA-GvHD) is caused by leukocytes, specifically T cells within a transfused blood product. Currently, the prevention of transfusion-associated graft-versus-host disease is performed by irradiation of blood products. With a sufficient reduction of leukocytes, the risk for TA-GvHD can be decreased. With consistent advances in current state-of-the-art blood filters, we herein propose that double filtration can sufficiently reduce leukocytes to reduce the risk for TA-GvHD. Materials Thirty RBC concentrates were filtered with leukocyte filters, followed by storage at 1–6 oC for 72 hours, and then a second filtration was performed. Residual leukocytes in the double-filtered RBC units (n = 30) were assessed with flow cytometric methods, and an additional assay with isolated peripheral blood mononuclear cells (PBMCs) (n = 6) was done by both flow cytometric methods and an automated hematology analyzer. Quality of the RBCs after filtration was evaluated by hematological and biochemical tests. In vitro T cell expansion was performed using anti-CD3/CD28-coated Dynabeads or anti-CD3 (OKT3). In vivo experiment for GvHD was performed by using NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice. Results Double-filtered blood products showed residual leukocyte levels below detection limits, which calculated to be below 1200–2500 cells per blood unit. In vitro expansion rate of T cells showed that 6x103 and 1x103 cell-seeded specimens showed 60.8±10.6 fold and 10.2±9.7-fold expansion, respectively. Cell expansion was not sufficiently observed in wells planted with 1x102 or 10 cells. In vivo experiments showed that mice injected with 1x105 or more cells cause fatal GvHD. GvHD induced inflammation was observed in mice injected with 1x104 or more cells. No evidence of GvHD was found in mice injected with 103 cells. Conclusions Our study suggests that additional removal of contaminating lymphocytes by a second leukodepletion step may further reduce the risk for TA-GvHD.
Collapse
Affiliation(s)
- Sejong Chun
- Department of Laboratory Medicine, Chonnam National University Medical School & Hospital, Gwangju, Korea
| | - Minh-Trang Thi Phan
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunwan University School of Medicine, Seoul, Korea
| | - Saetbyul Hong
- Animal Research and Molecular Imaging Center, Samsung Medical Center, Seoul, Korea
| | - Jehoon Yang
- Animal Research and Molecular Imaging Center, Samsung Medical Center, Seoul, Korea
| | - Yeup Yoon
- Animal Research and Molecular Imaging Center, Samsung Medical Center, Seoul, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunwan University School of Medicine, Seoul, Korea
| | - Sangbin Han
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jungwon Kang
- Blood Transfusion Research Institute, Korean Red Cross, Wonju, Korea
| | - Mark H. Yazer
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Jaehyun Kim
- Blood Transfusion Research Institute, Korean Red Cross, Wonju, Korea
- * E-mail: (DC); (JK)
| | - Duck Cho
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunwan University School of Medicine, Seoul, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunwan University School of Medicine, Seoul, Korea
- * E-mail: (DC); (JK)
| |
Collapse
|
10
|
徐 艳, 易 甜, 徐 肖, 裴 夫, 何 岳, 吴 学. [Effect of cyclophosphamide on hematopoietic stem cells in mice with iron overload]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:110-117. [PMID: 32376555 PMCID: PMC7040758 DOI: 10.12122/j.issn.1673-4254.2020.01.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To explore the effect of cyclophosphamide on hematopoietic stem cells (HSCs) in mice with iron overload. METHODS Mouse models of iron overload were established in 30 male C57BL/6 mice by intraperitoneal injections of iron dextran at low (0.25 g/kg), moderate (0.5 g/kg), and high (1 g/kg) doses (n=10), with another 10 PBS-treated mice as the control group. The changes in body weight, liver, spleen and bone marrow of the mice were recorded, and serum level of ferritin was detected. The mice receiving a moderate dose of iron dextran were further divided into 8 groups for observation at different time points (D1, D2, D3, D4, D5, D6, D7, and D14 groups) and were given intraperitoneal injection of 50 mg/kg cyclophosphamide (Cy) for 2 consecutive days. Peripheral blood cells, bone marrow mononuclear cells (BMMNCs), and the frequencies of different HSCs (HPCs, HSCs, LT-HSCs) in the BMMNCs were monitored. The cell cycle distribution in the HSCs, level of reactive oxygen species and the microenvironment of the HSCs were analyzed using flow cytometry. RESULTS Compared with the control mice, the mice with iron overload showed obvious weight loss with significantly increased serum ferritin level, enlargement of the liver and spleen, and iron deposition in the organs (P < 0.05). No significant changes were noted in the peripheral blood of the mice with iron overload. The cyclophosphamide-treated mice exhibited significantly decreased number of WBCs and lymphocyte ratio at days 1 to 4 (P < 0.05). The numbers of BMMNCs and HPCs in mice with iron overload did not show significant changes as compared with those in the control mice, but the numbers of HSCs and LTHSCs decreased significantly in the mice with iron overload (P < 0.05). In cyclophosphamide-treated mice, the number of HSCs increased since day 1 and reached the peak level on day 3 (P < 0.05). Compared with those in the control group, the HSCs did not exhibit significant changes in cell cycle distribution in mice with iron overload, but the proportion of G0/G1 cells decreased significantly in cyclophosphamide group since day 1 and reached the lowest level on day 3 (P < 0.05). CONCLUSIONS Iron deposition in the bone marrow causes long- term damages of the HSCs in the bone marrow but does not induce obvious changes in the peripheral blood. In mice with iron overload, intraperitoneal injection of 50 mg/kg cyclophosphamide for two days promotes cell cycle changes of the resting HSCs to mobilize the HSCs, and this effect is the most obvious on day 4.
Collapse
Affiliation(s)
- 艳军 徐
- 南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- 南方医科大学附属佛山妇幼保健院儿科,广东 佛山 528000Department of Pediatrics, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan 528000, China
| | - 甜甜 易
- 南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 肖肖 徐
- 南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 夫瑜 裴
- 南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 岳林 何
- 南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 学东 吴
- 南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
11
|
Chandra S, Cristofori P, Fonck C, O'Neill CA. Ex Vivo Gene Therapy: Graft-versus-host Disease (GVHD) in NSG™ (NOD.Cg-Prkdc scid Il2rg tm1Wjl/SzJ) Mice Transplanted with CD34 + Human Hematopoietic Stem Cells. Toxicol Pathol 2019; 47:656-660. [PMID: 31064282 DOI: 10.1177/0192623319844484] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A therapeutic option for monogenic disorders is gene therapy with ex vivo-transduced autologous hematopoietic stem cells (HSCs). Safety or efficacy studies of ex vivo-modified HSCs are conducted in humanized mouse models after ablation of the murine bone marrow and transfer of human CD34+ HSCs. Engrafted human CD34+ cells migrate to bone marrow and differentiate into various human hematopoietic lineages. A 12-week study was conducted in NSG™ mice to evaluate engraftment, differentiation, and safety of human CD34+ cells that were transduced (ex vivo) with a proprietary lentiviral vector encoding a human gene (BMRN-1) or a mock (green fluorescent protein) vector. Several mice intravenously injected with naive CD34+ cells or transduced CD34+ cells had variable lymphohistiocytic inflammatory cell infiltrates and microgranulomas in the liver and lungs consistent with graft-versus-host disease (GVHD). Spleen, bone marrow, stomach, reproductive tract, but not the skin had similar inflammatory changes. Ex vivo viral transduction of CD34+ cells did not impact engraftment or predispose to xenogeneic GVHD.
Collapse
Affiliation(s)
| | | | - Carlos Fonck
- 1 BioMarin Pharmaceutical Inc., San Rafael, California, USA
| | | |
Collapse
|
12
|
Wang K, Lv M, Chang YJ, Zhao XY, Zhao XS, Zhang YY, Sun YQ, Wang ZD, Suo P, Zhou Y, Liu D, Zhai SZ, Hong Y, Wang Y, Zhang XH, Xu LP, Liu KY, Huang XJ. Early myeloid-derived suppressor cells (HLA-DR -/ lowCD33 +CD16 -) expanded by granulocyte colony-stimulating factor prevent acute graft-versus-host disease (GVHD) in humanized mouse and might contribute to lower GVHD in patients post allo-HSCT. J Hematol Oncol 2019; 12:31. [PMID: 30885244 PMCID: PMC6423891 DOI: 10.1186/s13045-019-0710-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/21/2019] [Indexed: 02/06/2023] Open
Abstract
Introduction Myeloid-derived suppressor cells (MDSCs) are proposed to control graft-versus-host disease (GVHD) in allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, the definition of human MDSCs has not yet reached consensus, and the mechanism of MDSCs to control GVHD remains unclear. Methods Immature myeloid cells (HLA-DR−/lowCD33+CD16−) were tested before and after granulocyte colony-stimulating factor (G-CSF) administration in healthy donor and isolated for suppression assays and co-culture with T cells in vitro. Isolated cells were infused in humanized mice for a xenogeneic model of acute GVHD. One hundred allo-HSCT recipients were enrolled prospectively to assess the role of HLA-DR−/lowCD33+CD16− cells in grafts on the occurrence of acute GVHD. Results In the present study, G-CSF mobilized HLA-DR−/lowCD33+CD16− cells with immunosuppressive properties in donor peripheral blood. These cells contained more interleukin-10+ and transforming growth factor-beta (TGF-β)+ cells after G-CSF administration and inhibited the proliferation of autologous donor T cells in a TGF-β-dependent manner. Meanwhile, these immature myeloid cells promoted regulatory T cell expansion and induced Th2 differentiation. Importantly, these cells prevented acute GVHD in a humanized mouse model. Moreover, clinical cohort results showed that the number of HLA-DR−/lowCD33+CD16− cells in the donor graft was the only independent risk factor inversely correlated with the incidence of grade II–IV acute GVHD in the recipients (HR 0.388, 95% CI 0.158–0.954, p = 0.039). Conclusion HLA-DR−/lowCD33+CD16− cells represent functional MDSCs that may control acute GVHD in allo-HSCT. Electronic supplementary material The online version of this article (10.1186/s13045-019-0710-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ke Wang
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China.,Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, No.5 Yiheyuan Road, Beijing, 100871, China
| | - Meng Lv
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Ying-Jun Chang
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Xiao-Su Zhao
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Yuan-Yuan Zhang
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Yu-Qian Sun
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Zhi-Dong Wang
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Pan Suo
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Yang Zhou
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Dan Liu
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Shu-Zhen Zhai
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Yan Hong
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No 11 Xizhimen South Street, Beijing, 100044, China. .,Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, No.5 Yiheyuan Road, Beijing, 100871, China.
| |
Collapse
|
13
|
Huey DD, Bolon B, La Perle KMD, Kannian P, Jacobson S, Ratner L, Green PL, Niewiesk S. Role of Wild-type and Recombinant Human T-cell Leukemia Viruses in Lymphoproliferative Disease in Humanized NSG Mice. Comp Med 2018; 68:4-14. [PMID: 29460716 PMCID: PMC5824134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/29/2017] [Accepted: 05/30/2017] [Indexed: 06/08/2023]
Abstract
Chronic infection with human T-cell leukemia virus type 1 (HTLV1) can lead to adult T-cell leukemia (ATL). In contrast, infection with HTLV2 does not lead to leukemia, potentially because of distinct virus-host interactions and an active immune response that controls virus replication and, therefore, leukemia development. We created a humanized mouse model by injecting human umbilical-cord stem cells into the livers of immunodeficient neonatal NSG mice, resulting in the development of human lymphocytes that cannot mount an adaptive immune response. We used these mice to compare the ability of molecular clones of HTLV1, HTLV2, and select recombinant viruses to induce leukemia-lymphoma in vivo. Infection with HTLV1 strongly stimulated the proliferation of CD4+ T cells, whereas HTLV2 preferentially stimulated the proliferation of CD8+ T cells; both HTLV1 and HTLV2 induced lymphoproliferative disease. Uninfected and HTLV-infected humanized mice both showed granulomatous inflammation as a background lesion. Similarly, recombinant viruses that expressed the HTLV1 envelope protein (Env) on an HTLV2 background (HTLV2-Env1) or Env2 on an HTLV1 background (HTLV1-Env2) induced lymphoproliferative disease. HTLV2-Env1 stimulated the proliferation of CD4+ T cells, whereas HTLV1-Env2 stimulated both CD4+ and CD8+ T-cell subsets. Our results show that T-cell transformation in vivo is guided by the Env protein of the virus. Furthermore, our humanized mouse model is useful for exploring the preferred T-cell tropisms of HTLV1 and HTLV2.
Collapse
Affiliation(s)
- Devra D Huey
- Department of Veterinary Biosciences, Center for Retrovirus Research, College of Veterinary Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Brad Bolon
- Department of Veterinary Biosciences, Comparative Pathology and Mouse Phenotyping Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Krista M D La Perle
- Department of Veterinary Biosciences, Comparative Pathology and Mouse Phenotyping Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Priya Kannian
- Department of Veterinary Biosciences, Center for Retrovirus Research, College of Veterinary Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Steven Jacobson
- Neuroimmunology Branch, National Institute of Neurologic Disorders and Stroke, Bethesda, Maryland
| | - Lee Ratner
- Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Patrick L Green
- Department of Veterinary Biosciences, Center for Retrovirus Research, College of Veterinary Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, Center for Retrovirus Research, College of Veterinary Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH;,
| |
Collapse
|
14
|
Tashiro J, Rubio GA, Limper AH, Williams K, Elliot SJ, Ninou I, Aidinis V, Tzouvelekis A, Glassberg MK. Exploring Animal Models That Resemble Idiopathic Pulmonary Fibrosis. Front Med (Lausanne) 2017; 4:118. [PMID: 28804709 PMCID: PMC5532376 DOI: 10.3389/fmed.2017.00118] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/11/2017] [Indexed: 02/03/2023] Open
Abstract
Large multicenter clinical trials have led to two recently approved drugs for patients with idiopathic pulmonary fibrosis (IPF); yet, both of these therapies only slow disease progression and do not provide a definitive cure. Traditionally, preclinical trials have utilized mouse models of bleomycin (BLM)-induced pulmonary fibrosis—though several limitations prevent direct translation to human IPF. Spontaneous pulmonary fibrosis occurs in other animal species, including dogs, horses, donkeys, and cats. While the fibrotic lungs of these animals share many characteristics with lungs of patients with IPF, current veterinary classifications of fibrotic lung disease are not entirely equivalent. Additional studies that profile these examples of spontaneous fibroses in animals for similarities to human IPF should prove useful for both human and animal investigators. In the meantime, studies of BLM-induced fibrosis in aged male mice remain the most clinically relevant model for preclinical study for human IPF. Addressing issues such as time course of treatment, animal size and characteristics, clinically irrelevant treatment endpoints, and reproducibility of therapeutic outcomes will improve the current status of preclinical studies. Elucidating the mechanisms responsible for the development of fibrosis and disrepair associated with aging through a collaborative approach between researchers will promote the development of models that more accurately represent the realm of interstitial lung diseases in humans.
Collapse
Affiliation(s)
- Jun Tashiro
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Gustavo A Rubio
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Andrew H Limper
- Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Kurt Williams
- Department Pathobiology and Diagnostic Investigations, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Sharon J Elliot
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ioanna Ninou
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Vassilis Aidinis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Argyrios Tzouvelekis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Marilyn K Glassberg
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|