1
|
Skelin Ilic J, Bódi I, Milkovic L, Prodan Z, Belina D, Heckel D, Cicin-Sain L, Grčević D, Delfino DV, Radic Kristo D, Matulić M, Antica M. The Cellular and Molecular Characteristics of Postnatal Human Thymus Stromal Stem Cells. Biomedicines 2025; 13:1004. [PMID: 40299654 PMCID: PMC12024710 DOI: 10.3390/biomedicines13041004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/27/2025] [Accepted: 04/12/2025] [Indexed: 05/01/2025] Open
Abstract
Background: The thymus is the central hub of T-cell differentiation, where epithelial cells guide the process of their maturation. Objective: Our goal was to identify and describe progenitor cells within the human thymus that can differentiate into epithelial cells. Methods: When we plated enriched thymic cells in 3D culture conditions, rare individual cells capable of self-renewal and differentiation formed spheroids. Results: Both neonatal and adult thymuses produced similar numbers of spheroids, suggesting that progenitor potential remains consistent across age groups. Some cells within the spheres express genes typical of mature epithelial cells, while others express genes associated with the immature compartment active during thymic organogenesis. However, there were also cells expressing PDGFRβ. We treated the tissues with 2-deoxyguanosine before digestion, which improved the yield of progenitor cells. We also cultured the enriched stromal thymocytes with Cyr61 and Interleukin-22, which affected the spheroid size. Conclusions: Our efforts towards thymic reconstitution are ongoing, but our research uncovers previously unknown characteristics of the elusive epithelial progenitor population.
Collapse
Affiliation(s)
- Josipa Skelin Ilic
- Division of Molecular Mediicne, Rudjer Boskovic Institute, 10000 Zagreb, Croatia
| | - Ildikó Bódi
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1085 Budapest, Hungary
| | - Lidija Milkovic
- Division of Molecular Mediicne, Rudjer Boskovic Institute, 10000 Zagreb, Croatia
| | - Zsolt Prodan
- Kids Heart Center Budapest, 1096 Budapest, Hungary
| | - Dražen Belina
- Department of Cardiac Surgery, University Hospital Centre, 10000 Zagreb, Croatia
| | - Darko Heckel
- Division of Molecular Mediicne, Rudjer Boskovic Institute, 10000 Zagreb, Croatia
| | - Lipa Cicin-Sain
- Division of Molecular Mediicne, Rudjer Boskovic Institute, 10000 Zagreb, Croatia
| | - Danka Grčević
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Domenico Vittorio Delfino
- Foligno Nursing School, Department of Medicine and Surgery, University of Perugia, 06034 Foligno, Italy
- Section of Pharmacology, Department of Medicine, University of Perugia, 06123 Perugia, Italy
| | | | - Maja Matulić
- Department of Biology, Faculty of Science, 10000 Zagreb, Croatia
| | - Mariastefania Antica
- Division of Molecular Mediicne, Rudjer Boskovic Institute, 10000 Zagreb, Croatia
- Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| |
Collapse
|
2
|
Transplantation tolerance induced in humans at the fetal or the neonatal stage. J Transplant 2011; 2011:760319. [PMID: 21876781 PMCID: PMC3159006 DOI: 10.1155/2011/760319] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 05/06/2011] [Indexed: 11/30/2022] Open
Abstract
Patients transplanted with HLA-mismatched stem cells from fetal livers develop transplantation tolerance to donor antigens. Engraftment needs no conditioning regimen prior to transplantation in neonates with severe combined immunodeficiency disease or in human fetal patients having not yet developed any immune maturity, especially T-cell differentiation. The chimeric patients have donor-derived T lymphocytes which progressively demonstrate positive interactions with other host cells. They also can be shown to be tolerant toward both host and donor antigens. The latter tolerance relies upon clonal deletion from the T-cell repertoire, and it results from the contact between thymocytes of donor origin and dendritic cells or macrophages also deriving from donor stem cells. The former tolerance does not imply clonal deletion of T-cells with host reactivity. Numerous T-cells recognizing the allogeneic, host-type antigens are identified in these patients, but these cells are anergized, following interaction with epithelial cells of the host thymus. Induction of transplantation tolerance at the fetal stage requires minimal engraftment only; in the future it will be possible to further amplify the clinical benefit, using additional cell transplants after birth.
Collapse
|
3
|
Joo SY, Choi BK, Kang MJ, Jung DY, Park KS, Park JB, Choi GS, Joh J, Kwon CH, Jung GO, Lee SK, Kim SJ. Development of functional human immune system with the transplantations of human fetal liver/thymus tissues and expanded hematopoietic stem cells in RAG2-/-gamma(c)-/- MICE. Transplant Proc 2009; 41:1885-90. [PMID: 19545750 DOI: 10.1016/j.transproceed.2009.02.074] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 12/03/2008] [Accepted: 02/09/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND There is an increasing need for suitable animal models for the study of the human immune system and disease. The purpose of this study was to develop a practical in vivo model of human immune cell repopulation using ex vivo expanded human fetal liver-derived CD34(+) hematopoietic stem cells and subrenally coimplanted fetal liver/thymus tissues. METHODS Freshly isolated fetal liver-derived CD34(+) hematopoietic stem cells were frozen until injected and ex vivo expanded with various cytokines for 7 days. After fetal liver/thymus tissues were subrenally coimplanted into preirradiated Rag2(-/-)gamma(c)(-/-) mice, frozen and ex vivo expanded CD34(+) cells were injected intravenously. The peripheral blood of the mice was monitored for the detection of human cell engraftment using flow cytometry. Then we confirmed human T-cell function by in vitro function assays. RESULTS After fetal liver/thymus tissues were coimplanted into the irradiated Rag2(-/-)gamma(c)(-/-) mice, with frozen and ex vivo expanded CD34(+) hematopoietic stem cells, human cell engraftments were determined using hCD45 and multilineage markers. The cultured cells with the cytokine combination of stem cell factor, thrombopoietin, Flk2/Flk3 ligand (FL), and interleukin-3 showed stable and long-term engraftment compared to other combinations. The ex vivo expanded human fetal liver-derived CD34(+) hematopoietic stem cells, under our culture conditions, accomplished a large volume of expanded cells that were sustained, demonstrating self-renewal of the evaluated markers, which may have indicated long- term repopulation activity. CONCLUSION The results of this study demonstrated a practical mouse model of expanded human immune cells especially T cells in Rag2(-/-)gamma(c)(-/-) mice.
Collapse
Affiliation(s)
- S-Y Joo
- Transplantation Research Center, Samsung Biomedical Research Institute, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Roncarolo MG, Carballido JM. Construction of human-SCID chimeric mice. ACTA ACUST UNITED AC 2008; Chapter 4:4.8.1-4.8.17. [PMID: 18432798 DOI: 10.1002/0471142735.im0408s25] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Until recently, testing of new therapeutic agents has relied extensively upon the use of mice and nonhuman primates for in vivo preclinical studies. Unfortunately, these animal models do not always mimic the physiological and pathophysiological processes that occur in humans. The finding that C.B-17 severe combined immunodeficiency (SCID) mice lack a competent immune system, and therefore are unable to mount effective cellular and humoral responses to foreign antigens, has led to their use as recipients for xenografts of human tissues. This unit is focused on the construction of human-SCID chimeric through the surgical implantation of human fetal hematolymphoid tissues into SCID mice (SCID-hu mice). The Basic Protocol describes the surgical implantation of human fetal thymus and liver under the kidney capsules of SCID mice (SCID-hu Thy/Liv model). Subcutaneous transplantation of human fetal bone marrow and thymus (SCID-hu Bm/Thy mice) is described in the Alternate Protocol. Additional support protocols provide procedures to analyze human lymphocyte populations in the peripheral blood and grafted organs of SCID-hu mice. The advantages and disadvantages of each protocol and potential applications are discussed in the Commentary.
Collapse
|
5
|
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: 44] [Impact Index Per Article: 2.6] [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
|
6
|
Abstract
Inbred mice with specific genetic defects have greatly facilitated the analysis of complex biological events. Several humanized mouse models using the C.B.-17 scid/scid mouse (referred to as the SCID mouse) have been created from two transplantation protocols, and these mice have been utilized for the investigation of human immunodeficiency virus type 1 (HIV-1) and human T-lymphotropic virus type I (HTLV-I) pathogenesis and the evaluation of antiviral compounds. To generate a more prominent small animal model for human retrovirus infection, especially for examination of the pathological process and the immune reaction, a novel immunodeficient mouse strain derived from the NOD SCID mouse was created by backcrossing with a common gamma chain (gamma(c))-knockout mouse. The NOD-SCID gamma(c)null (NOG) mouse has neither functional T and B cells nor NK cells and has been used as a recipient in humanized mouse models for transplantation of human immune cells particularly including hematopoietic stem cells (HSC). From recent advances in development of humanized mice, we are now able to provide a new version of the animal model for human retrovirus infection and human immunity.
Collapse
|
7
|
Nonprimate models of HIV-1 infection and pathogenesis. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2008; 56:399-422. [PMID: 18086419 DOI: 10.1016/s1054-3589(07)56013-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
8
|
Touraine JL, Plotnicky H, Roncarolo MG, Bacchetta R, Gebuhrer L. Immunological lessons learnt from patients transplanted with fully mismatched stem cells. Immunol Res 2007; 38:201-9. [PMID: 17917026 DOI: 10.1007/s12026-007-0002-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/29/2022]
Abstract
Fully HLA-mismatched stem cells from human fetal livers were transplanted into 17 infants and two fetuses to treat severe combined immunodeficiency disease in 1976-2000. Donor cell engraftment and immunological reconstitution were obtained in 14/19 patients, three of whom have been extensively and repeatedly studied immunologically during prolonged follow-up. T-cells were derived totally from donor cells; B-cells and antigen-presenting cells (APC) remained mainly of host origin. Due to class I and II mismatches between T-cells and all other cells (APC, B-cells, virus-infected target cells), limitations in the defense against infections in vivo and in T-cell functions in vitro (helper and cytotoxic activities) were predicted; however, these did not occur. Anti-tetanus toxoid responses (including specific antibody production) developed despite HLA disparities between T-cells and B-cells or APC in the chimeric children. Similarly, cytotoxic T-cells (of donor HLA phenotype) recognized host Epstein-Barr virus-infected target cells. Recognition of antigenic peptide by T-cells under these conditions involved presentation by host allogeneic HLA molecules and not by self HLA antigens. Tolerance to donor antigens was acquired by clonal deletion; tolerance to host antigens existed despite the presence of many host-reactive T-cells and involved clonal anergy.
Collapse
Affiliation(s)
- Jean-Louis Touraine
- Department of Transplantation and Clinical Immunology, Claude Bernard University and Edouard Herriot Hospital, Lyon, France.
| | | | | | | | | |
Collapse
|
9
|
Abstract
BACKGROUND Angiogenesis and tumor-associated immunosuppression are two of the hallmarks of carcinogenesis. In previous studies we demonstrated in vitro that HNSCC tumor cells attract monocytes via monocyte chemotactic protein-1 (MCP-1) and activate them via transforming growth factor-beta 1(TGF-beta1) to secrete interleukin (IL)-1alpha, which in turn stimulates tumor cells to secrete increased levels of the angiogenic and immunosuppressive vascular endothelial growth factor (VEGF). These findings suggest that interaction between the immune system and VEGF-mediated angiogenesis is important for progression of HNSCC. Recent studies in vitro show that retinoic acid (RA) downregulates the release of MCP-1 and TGF-beta1 by tumor cells. Therefore, we investigated the ability of RA to modulate the ability of tumor cells to recruit and activate monocytes for participation in VEGF-mediated angiogenesis and immunosuppression in vivo. MATERIAL AND METHODS Mice (ten/group) were injected daily with RA (160 microg/kg) for 3 weeks. After that time mice were sacrificed, and paraffin sections of tumors were obtained and stained for VEGF-A, CD68, and PECAM (CD31) by immunohistochemistry. The lungs, liver, and myocardium were analyzed for macro- and micrometastases. The plasma protein levels of VEGF-A and MCP-1 were determined by ELISA. RESULTS In RA-treated mice tumors regressed completely and RA prevented metastases (p=0.00) and macrophage infiltration (p=0.007). Treated mice downregulated VEGF-A (0 pg/ml) and MCP-1 (12 pg/ml) in peripheral blood (p=0.001). CONCLUSION Our findings suggest a new therapeutic possibility: the development of treatment protocols that can block each of the ways in which tumors induce new blood vessel growth and immunosuppression of the host.
Collapse
Affiliation(s)
- L Strauss
- Abteilung Onkologie, Universitätsspital Zürich.
| | | | | | | |
Collapse
|
10
|
Abstract
How HIV replicates and causes destruction of the thymus, and how to restore thymic function, are among the most important questions of HIV-1 pathogenesis and therapy in adult as well as pediatric patients. The thymus appears to function, albeit at reduced levels, throughout the life of adults, to respond to T cell depletion induced by HIV and to be suppressed by HIV. In this review, we summarize recent findings concerning HIV replication and pathogenesis in the human thymus, focusing on mechanistic insights gleaned from studies in the SCID-hu Thy/Liv mouse and human fetal-thymus organ culture (HF-TOC) models. First, we discuss HIV viral determinants and host factors involved in the replication of HIV in the thymus. Second, we consider evidence that both viral factors and host factors contribute to HIV-induced thymocyte depletion. We thus propose that multiple mechanisms, including depletion and suppression of progenitor cells, paracrine and direct lytic depletion of thymocytes, and altered thymocyte selection are involved in HIV-induced pathology in the thymus. With the SCID-hu Thy/Liv mouse and HF-TOC models, it will be important in the coming years to further clarify the virological, cell biological, and immunological mechanisms of HIV replication and pathogenesis in human thymus, and to correlate their significance in HIV disease progression.
Collapse
Affiliation(s)
- Eric G Meissner
- Department of Microbiology and Immunology, The Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina, Chapel Hill, NC 27599-7295, USA
| | | | | | | | | |
Collapse
|
11
|
Abstract
The thymus is the primary site of T-cell production early in life, and has now been shown to continue to function in both healthy and immunocompromised individuals late into life. Positive and negative selection occurring in the thymus are two of the most important processes that govern the development and specificity of peripheral T cells, including their restriction to self HLA and their ability to respond in an alloreactive manner. In the chimeric state that follows successful allogeneic stem-cell transplants, the specificity of alloreactive cells may be governed by either host- or recipient-derived cellular elements, as well as maturing lymphoid cells, which are, in turn, derived from donor stem cells or host cells surviving transplant conditioning. The ability to measure recent thymic emigrants via the detection of T-cell receptor excision circles has facilitated studies of thymic function in immunodeficient individuals, including HIV-1 infected subjects and recipients of autologous or allogeneic stem-cell transplant (SCT). These studies have now demonstrated that thymic function is likely to play a beneficial role in immune reconstitution in these settings, but have yet to clearly demonstrate what clinical variables are the most important determinants of thymic persistence. It is also not yet clear how much the degree of thymic function following allogeneic SCT influences the alloreactive T-cell repertoire, although studies in animal models and early clinical studies suggest that GvHD results in thymic injury and dysfunction. Future studies will further clarify how thymic function shapes the repertoire of T cells that mediate alloreactivity, as well as protective pathogen-specific immune responses, following SCT. Finally, these studies will also demonstrate whether endogenous mediators of thymic function could be selectively applied to regulate post-SCT thymic function and alloreactivity.
Collapse
Affiliation(s)
- K V Komanduri
- Department of Blood and Marrow Transplantation, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
12
|
Abstract
The thymus is the main producer of alphabeta T cells and is, therefore, crucial for a normal immune system. The intrathymic developmental pathway of human alphabeta T cells has now been delineated. The production of new T cells by the thymus decreases with age, and the thymus was thought to be redundant in adults once the peripheral T-cell pool has been formed early in life. However, recent work has shown that the thymus can function even at an advanced age. Research into the production of T cells in clinical settings that are associated with loss of T cells in the periphery has sparked renewed interest in the function of the human thymus.
Collapse
Affiliation(s)
- Hergen Spits
- Department of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
13
|
Yahata T, Ando K, Nakamura Y, Ueyama Y, Shimamura K, Tamaoki N, Kato S, Hotta T. Functional human T lymphocyte development from cord blood CD34+ cells in nonobese diabetic/Shi-scid, IL-2 receptor gamma null mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:204-9. [PMID: 12077246 DOI: 10.4049/jimmunol.169.1.204] [Citation(s) in RCA: 152] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
An experimental model for human T lymphocyte development from hemopoietic stem cells is necessary to study the complex processes of T cell differentiation in vivo. In this study, we report a newly developed nonobese diabetic (NOD)/Shi-scid, IL-2Rgamma null (NOD/SCID/gamma(c)(null)) mouse model for human T lymphopoiesis. When these mice were transplanted with human cord blood CD34(+) cells, the mice reproductively developed human T cells in their thymus and migrated into peripheral lymphoid organs. Furthermore, these T cells bear polyclonal TCR-alphabeta, and respond not only to mitogenic stimuli, such as PHA and IL-2, but to allogenic human cells. These results indicate that functional human T lymphocytes can be reconstituted from CD34(+) cells in NOD/SCID/gamma(c)(null) mice. This newly developed mouse model is expected to become a useful tool for the analysis of human T lymphopoiesis and immune response, and an animal model for studying T lymphotropic viral infections, such as HIV.
Collapse
MESH Headings
- Animals
- Antigens, CD34/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/analysis
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cells, Cultured
- Crosses, Genetic
- Fetal Blood/cytology
- Fetal Blood/immunology
- Fetal Blood/metabolism
- Humans
- Immunophenotyping
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Receptors, Interleukin-2/deficiency
- Receptors, Interleukin-2/genetics
- Species Specificity
- Spleen/cytology
- Spleen/immunology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- Thymus Gland/cytology
- Thymus Gland/immunology
Collapse
Affiliation(s)
- Takashi Yahata
- Research Center for Cell Transplantation, Kanagawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Denaro M, Kolber-Simonds D, Schad V, Muthukumar S, Germana S, White-Scharf ME, Banerjee PT, LeGuern C, Andersson G. Expression of xenogeneic MHC class II molecules in HLA-DR(+) and -DR(-) cells: influence of retrovirus vector design and cellular context. Xenotransplantation 2002; 9:115-24. [PMID: 11897004 DOI: 10.1034/j.1399-3089.2002.1o038.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We recently established that molecular chimeras of major histocompatibility complex (MHC) class II molecules, created via retroviral transfer of allogeneic class II cDNAs into bone marrow cells (BMCs), alleviated complications associated with mixed BMC chimeras while leading to T cell tolerance to renal grafts sharing the transferred class II. Initially demonstrated for allogeneic transplants in miniature swine, this concept was extended to T-dependent antibody (Ab) responses to xenogeneic antigens (Ags) in the pig --> baboon combination. Successful down-regulation of T cell responses appeared, however, to be contingent on a tight lineage-specific expression of transferred class II molecules. The present studies were, therefore, designed to evaluate the influence of construct design and cellular environment on expression of retrovirally transferred xenogeneic class II cDNAs. Proviral genomes for pig class II SLA-DR expression, differing only at the marker neo(r) or enhanced green fluorescent protein (EGFP) gene, showed increased membrane SLA-DR density on HLA-DR(-) fibroblasts as well as HLA-DR(+), TF-1 erythroleukemia cells. More importantly, HLA-DR(+) human B cell lines, although efficiently transduced with pig DR retroviruses, exhibited unstable surface pig DR. Surface pig DR- B cells, nevertheless, stimulated autologous human T cells pre-sensitized to pig Ags, a proliferation likely occurring through presentation of class II-derived peptides. Collectively, these data suggest that surface expression of transferred class II molecules is not related to the ability of recipient cells to synthesize xenogeneic class II molecules but rather to their Ag processing capacities.
Collapse
Affiliation(s)
- Maria Denaro
- BioTransplant Incorporated, Charlestown Navy Yard, Charlestown, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Austin TW, Moss K, Morel F, Plavec I. An improved vector for high-level, consistent retroviral transgene expression in human thymocytes after competitive reconstitution from transduced peripheral blood stem cells. Hum Gene Ther 2001; 12:1239-49. [PMID: 11440618 DOI: 10.1089/104303401750270904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
One problem in hematopoietic stem cell (HSC)-based gene therapy is the low-level, and often transient, transgene expression in progeny cells in vivo. Here we have evaluated retroviral vector designs for improved long-term in vivo transgene expression levels in thymocytes recovered after transplantation of gene-modified HSCs. First, several vector designs were screened in vitro by single-cell analysis of transgene marking and expression to rapidly identify optimal vectors for sensitive tracking of marked cells. Next, using one optimal vector, we show that gene-modified HSCs can competitively reconstitute thymopoiesis in SCID-hu thymus/liver mice, with transgene expression detectable on 0-40% of marked donor thymocytes. Modified vector designs (termed MSCV-SAR and MoMLV-SAR), which enhance transgene expression in primary T cells in vitro, were shown here to improve in vivo transgene expression levels per cell 12- to 14-fold (mean fluorescence intensity was 2175 for MSCV-SAR vs. 174 for LNGFRSN; %NGFR(+) donor(+) cells with high-level expression was 58% for MSCV-SAR vs. 4% for LNGFRSN). Importantly, 61% of grafts had high-level transgene expression on thymocytes with the MSCV-SAR vector versus 0% of grafts for LNGFRSN or MoMLV-SAR. Transgene expression was demonstrated in various stages of thymocyte differentiation and was consistently detected in early thymic progenitors. We suggest that the MSCV-SAR vector described here is particularly advantageous for applications requiring high-level, consistent transgene expression in a diverse repertoire of T cells derived from gene-modified HSC grafts.
Collapse
Affiliation(s)
- T W Austin
- SyStemix, Inc., A Novartis Company, Palo Alto, CA 94304, USA.
| | | | | | | |
Collapse
|
16
|
Affiliation(s)
- M G Roncarolo
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy.
| | | | | |
Collapse
|
17
|
Shih CC, Hu J, Arber D, LeBon T, Forman SJ. Transplantation and growth characteristics of human fetal lymph node in immunodeficient mice. Exp Hematol 2000; 28:1046-53. [PMID: 11008017 DOI: 10.1016/s0301-472x(00)00518-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The lymph node is an integral component of the immune system and the major site of antigen-dependent lymphocyte proliferation and differentiation. Development of animal models possessing functional primary human lymph nodes will have a significant impact on research in lymphopoiesis and immune response. To date, successful transplantation of primary human lymph nodes in rodents has not yet been reported. This work was undertaken to develop a reliable methodology to engraft primary human fetal lymph nodes in immunodeficient mice. MATERIALS AND METHODS Three different sets of parameters, including three different transplantation sites in the mice, two different strains of immunodeficient mice, and two different preconditioning regimens, were evaluated. The growth characteristics of the implanted primary human fetal lymph nodes were examined 3 months after transplantation by histologic, immunocytochemical, and flow cytometric methods. RESULTS Transplantation of primary human fetal lymph nodes into subcutaneous pouches in the ears in severe combined immunodeficiency (SCID) mice preconditioned with etoposide reproducibly give rise to >80% engraftment. The engrafted primary human fetal lymph nodes undergo massive growth (>200-fold) and retain the same histology and cellular composition as fresh human fetal lymph nodes from the same donors. CONCLUSIONS We report, for the first time, the development of a reliable methodology to successfully engraft human fetal lymph node in SCID mice. The engrafted human lymph nodes are visible and accessible to experimental manipulations. This SCID-hu mouse model with human lymph node should provide a physiologically relevant system to investigate lymphopoiesis, immunologic response, and virus-mediated immunosuppression.
Collapse
Affiliation(s)
- C C Shih
- Division of Hematology/Bone Marrow Transplantation, City of Hope National Medical Center, Duarte, Calif., USA.
| | | | | | | | | |
Collapse
|
18
|
Salmon H, Johnson I, Germana S, Haller GW, Sachs DH, Leguern C. Dendritic cells enriched from swine thymus co-express CD1, CD2 and major histocompatibility complex class II and actively stimulate alloreactive T lymphocytes. Scand J Immunol 2000; 52:164-72. [PMID: 10931384 DOI: 10.1046/j.1365-3083.2000.00768.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Initial characterization and partial purification of thymic dendritic cells (DC) from miniature swine were carried out with the ultimate goal of using these cells to induce transplantation tolerance in this preclinical animal model. Immunohistochemical analysis of swine thymic tissue sections has shown DC to be large cells located in the medullary and the cortico-medullary regions as evidenced by the presence of surrounding Hassal bodies. These cells exhibit membrane processes and express the CD1, granulocyte/macrophage (G/M), and major histocompatibility complex (MHC) class II surface antigens, as well as the S100 cytosolic and nuclear markers found in humans to be specific for DC. Dendritic cells were purified from thymi following collagenase treatment, Percoll gradient centrifugation, and adhesion steps to plastic. Cells similar in morphology and phenotype to those described in tissue sections were detected in the lighter density layers of the gradient and represented 0.02% of the starting cell number. Removal of plastic nonadherent cells showed enrichment levels similar to those reported for murine and human DC. Two-colour flow cytometric analysis of purified pig DC identified these cells as MHC class IIhi, CD1+, CD2+, and G/M+. The dendritic nature of these cells was confirmed by their potent ability to stimulate alloreactive T lymphocytes. Modification of porcine thymic DC by transfer of allogeneic MHC genes and reinjection into the DC donor should permit testing of the role of this DC subset in the induction of transplantation tolerance.
Collapse
Affiliation(s)
- H Salmon
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, MGH-East, Bldg 149-9019, 13th Street, Boston, MA 01229, USA
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
Abstract
Thymic repopulation by transplanted hematopoietic progenitor cells (HPC) is likely to be important for long-term immune reconstitution and for successful gene therapy of diseases affecting the T-cell lineage. However, the T-cell progenitor potential of HPC, cultured in vitro for cell number expansion and gene transfer remains largely unknown. Here, we cultured highly purified human umbilical cord blood (CB) CD34+CD38− or CD34+CD38+ cells for up to 5 weeks in stroma-free cultures supplemented with various combinations of the cytokines thrombopoietin (TPO), stem cell factor (SCF), flt3/flk-2 ligand (FL), interleukin-3 (IL-3), and IL-6 and investigated thymus-repopulating ability of expanded cells in vitro and in vivo. After up to 5 weeks of culture in IL-3 + SCF + IL-6 or TPO + FL + SCF supplemented medium, the progeny of CD34+CD38− CB cells generated T cells and natural killer cells in the thymus. Limiting dilution experiments demonstrated increase in the number of T-cell progenitors during culture. After 3 weeks of culture, gene marked CD34+CD38− CB cells injected in the human thymus fragment transplanted in severe combined immunodeficient (SCID) mice (SCID-hu) generated thymocytes expressing the retroviral encoded marker gene GFP in vivo. Thus, our results show that the progeny of CD34+CD38− CB cells cultured for extensive periods, harbor thymus-repopulating cells that retain T-cell progenitor potential after expansion and gene transfer.
Collapse
|
20
|
Thymic Repopulation by CD34+ Human Cord Blood Cells After Expansion in Stroma-Free Culture. Blood 1999. [DOI: 10.1182/blood.v94.11.3644.423k06_3644_3652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Thymic repopulation by transplanted hematopoietic progenitor cells (HPC) is likely to be important for long-term immune reconstitution and for successful gene therapy of diseases affecting the T-cell lineage. However, the T-cell progenitor potential of HPC, cultured in vitro for cell number expansion and gene transfer remains largely unknown. Here, we cultured highly purified human umbilical cord blood (CB) CD34+CD38− or CD34+CD38+ cells for up to 5 weeks in stroma-free cultures supplemented with various combinations of the cytokines thrombopoietin (TPO), stem cell factor (SCF), flt3/flk-2 ligand (FL), interleukin-3 (IL-3), and IL-6 and investigated thymus-repopulating ability of expanded cells in vitro and in vivo. After up to 5 weeks of culture in IL-3 + SCF + IL-6 or TPO + FL + SCF supplemented medium, the progeny of CD34+CD38− CB cells generated T cells and natural killer cells in the thymus. Limiting dilution experiments demonstrated increase in the number of T-cell progenitors during culture. After 3 weeks of culture, gene marked CD34+CD38− CB cells injected in the human thymus fragment transplanted in severe combined immunodeficient (SCID) mice (SCID-hu) generated thymocytes expressing the retroviral encoded marker gene GFP in vivo. Thus, our results show that the progeny of CD34+CD38− CB cells cultured for extensive periods, harbor thymus-repopulating cells that retain T-cell progenitor potential after expansion and gene transfer.
Collapse
|
21
|
Touraine JL. Induction of transplantation tolerance in humans using stem cell transplants prenatally or postnatally. Transplant Proc 1999; 31:2735-7. [PMID: 10578269 DOI: 10.1016/s0041-1345(99)00545-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- J L Touraine
- Department of Transplantation and Clinical Immunology, E. Herriot Hospital, Lyon, France
| |
Collapse
|
22
|
Kovalev G, Duus K, Wang L, Lee R, Bonyhadi M, Ho D, McCune JM, Kaneshima H, Su L. Induction of MHC Class I Expression on Immature Thymocytes in HIV-1-Infected SCID-hu Thy/Liv Mice: Evidence of Indirect Mechanisms. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.12.7555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
The SCID-hu Thy/Liv mouse and human fetal thymic organ culture (HF-TOC) models have been used to explore the pathophysiologic mechanisms of HIV-1 infection in the thymus. We report here that HIV-1 infection of the SCID-hu Thy/Liv mouse leads to the induction of MHC class I (MHCI) expression on CD4+CD8+ (DP) thymocytes, which normally express low levels of MHCI. Induction of MHCI on DP thymocytes in HIV-1-infected Thy/Liv organs precedes their depletion and correlates with the pathogenic activity of the HIV-1 isolates. Both MHCI protein and mRNA are induced in thymocytes from HIV-1-infected Thy/Liv organs, indicating induction of MHCI gene expression. Indirect mechanisms are involved, because only a fraction (<10%) of the DP thymocytes were directly infected by HIV-1, although the majority of DP thymocytes are induced to express high levels of MHCI. We further demonstrate that IL-10 is induced in HIV-1-infected thymus organs. Similar HIV-1-mediated induction of MHCI expression was observed in HF-TOC assays. Exogenous IL-10 in HF-TOC induces MHCI expression on DP thymocytes. Therefore, HIV-1 infection of the thymus organ leads to induction of MHCI expression on immature thymocytes via indirect mechanisms involving IL-10. Overexpression of MHCI on DP thymocytes can interfere with thymocyte maturation and may contribute to HIV-1-induced thymocyte depletion.
Collapse
Affiliation(s)
- Grigoriy Kovalev
- *Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599
| | - Karen Duus
- *Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599
| | - Liping Wang
- *Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599
| | - Robert Lee
- †SyStemix, Inc., Palo Alto, CA 94304; and
| | | | - David Ho
- ‡Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016
| | | | | | - Lishan Su
- *Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
23
|
Kovalev G, Duus K, Wang L, Lee R, Bonyhadi M, Ho D, McCune JM, Kaneshima H, Su L. Induction of MHC class I expression on immature thymocytes in HIV-1-infected SCID-hu Thy/Liv mice: evidence of indirect mechanisms. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 1999; 162:7555-62. [PMID: 10358212 PMCID: PMC4435947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
The SCID-hu Thy/Liv mouse and human fetal thymic organ culture (HF-TOC) models have been used to explore the pathophysiologic mechanisms of HIV-1 infection in the thymus. We report here that HIV-1 infection of the SCID-hu Thy/Liv mouse leads to the induction of MHC class I (MHCI) expression on CD4+CD8+ (DP) thymocytes, which normally express low levels of MHCI. Induction of MHCI on DP thymocytes in HIV-1-infected Thy/Liv organs precedes their depletion and correlates with the pathogenic activity of the HIV-1 isolates. Both MHCI protein and mRNA are induced in thymocytes from HIV-1-infected Thy/Liv organs, indicating induction of MHCI gene expression. Indirect mechanisms are involved, because only a fraction (<10%) of the DP thymocytes were directly infected by HIV-1, although the majority of DP thymocytes are induced to express high levels of MHCI. We further demonstrate that IL-10 is induced in HIV-1-infected thymus organs. Similar HIV-1-mediated induction of MHCI expression was observed in HF-TOC assays. Exogenous IL-10 in HF-TOC induces MHCI expression on DP thymocytes. Therefore, HIV-1 infection of the thymus organ leads to induction of MHCI expression on immature thymocytes via indirect mechanisms involving IL-10. Overexpression of MHCI on DP thymocytes can interfere with thymocyte maturation and may contribute to HIV-1-induced thymocyte depletion.
Collapse
Affiliation(s)
- G Kovalev
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, University of North Carolina, Chapel Hill 27599, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Nikolic B, Gardner JP, Scadden DT, Arn JS, Sachs DH, Sykes M. Normal Development in Porcine Thymus Grafts and Specific Tolerance of Human T Cells to Porcine Donor MHC. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.6.3402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The induction of T cell tolerance is likely to play an essential role in successful xenotransplantation in humans. In this study, we show that porcine thymus grafts in immunodeficient mice support normal development of polyclonal, functional human T cells. These T cells were specifically tolerant to MHC Ags of the porcine thymus donor and responded to nondonor porcine xenoantigens and alloantigens. Exogenous IL-2 did not abolish tolerance, suggesting central clonal deletion rather than anergy as the likely tolerance mechanism. Our study suggests that the thymic transplantation approach to achieving tolerance with restoration of immunocompetence may be applicable to xenotransplantation of pig tissues to humans.
Collapse
Affiliation(s)
| | - Jason P. Gardner
- †Division of Experimental Hematology, AIDS Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - David T. Scadden
- †Division of Experimental Hematology, AIDS Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | | | | | - Megan Sykes
- *Transplantation Biology Research Center and
| |
Collapse
|
25
|
Péault B, Touraine JL, Charbord P. Haematopoietic stem cell emergence and development in the human embryo and fetus; perspectives for blood cell therapies in utero. ACTA ACUST UNITED AC 1999. [DOI: 10.1016/s1084-2756(99)80007-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
26
|
Touraine JL, Raudrant D, Laplace S, Gebuhrer L. Stem cell transplants in utero for genetic diseases: treatment and a model for induction of immunologic tolerance. Transplant Proc 1999; 31:681-2. [PMID: 10083293 DOI: 10.1016/s0041-1345(98)01606-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- J L Touraine
- Department of Transplantation and Clinical Immunology, Hôpital E. Herriot, Lyon, France
| | | | | | | |
Collapse
|
27
|
Kinoshita S, Chen BK, Kaneshima H, Nolan GP. Host control of HIV-1 parasitism in T cells by the nuclear factor of activated T cells. Cell 1998; 95:595-604. [PMID: 9845362 DOI: 10.1016/s0092-8674(00)81630-x] [Citation(s) in RCA: 186] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Post HIV-1 entry, productive HIV-1 infection of primary T cells requires overcoming several cellular blocks to provirus establishment and replication. Activation of unknown host intracellular events overcomes such inhibitory steps and is concomitant with HIV-1 replication. We show that the transcription factor NFATc was sufficient as a cellular factor to induce a highly permissive state for HIV-1 replication in primary CD4+ T cells. NFATc overcame a blockade at reverse transcription and permitted active HIV-1 replication. Pharmacologic blockade of endogenous NFAT activity by FK506 or CsA inhibited synthesis of reverse transcription and also potently blocked HIV-1 replication. T cells therefore can become competent for HIV-1 replication by control of regulated host factors such as the NFATc transcription factor. The host mechanisms regulated by such permissivity factors are potential targets for anti-HIV-1 therapy.
Collapse
Affiliation(s)
- S Kinoshita
- Department of Molecular Pharmacology, Stanford University School of Medicine, California 94305, USA
| | | | | | | |
Collapse
|
28
|
Berkowitz RD, Beckerman KP, Schall TJ, McCune JM. CXCR4 and CCR5 Expression Delineates Targets for HIV-1 Disruption of T Cell Differentiation. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.7.3702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
HIV-1 disease is often associated with CD4+ T lymphopenia as well as quantitative reductions in naive CD8+ T cells and cytopenias involving nonlymphoid hemopoietic lineages. Studies in HIV-1-infected humans as well as in animal models of lentivirus disease indicate that these effects may be secondary to infection and destruction of multilineage and lineage-restricted hemopoietic progenitor cells. To define the stages of T cell differentiation that might be susceptible to HIV-1, we performed flow cytometric analysis of the surface expression of CXCR4 and CCR5 on T cells and their progenitors from fetal tissue, cord blood, SCID-hu Thy/Liv mice, and adult peripheral blood. We found that CXCR4 is expressed at low levels on hemopoietic progenitors in the bone marrow, is highly expressed on immature (CD3−CD4+CD8−) T cell progenitors in the thymus, and then is down-regulated during thymocyte differentiation. As thymocytes leave the thymus and enter the peripheral circulation, the expression of CXCR4 is again up-regulated. In contrast, CCR5 is undetectable on most hemopoietic progenitors in the bone marrow and on intrathymic T progenitor cells. It is up-regulated when thymocytes coexpress CD4 and CD8, then down-regulated either in the thymus (CD4+ cells) or during exit from the thymus (CD8+ cells). These results indicate that discrete, lineage-related populations of T cell progenitors may vary widely in their potential to respond to chemokines and to be infected by HIV-1, and that T lymphoid differentiation is particularly vulnerable to CXCR4-using viruses.
Collapse
Affiliation(s)
- Robert D. Berkowitz
- *Gladstone Institute of Virology and Immunology, San Francisco, CA 94141, and Departments of Microbiology and Immunology and Medicine, University of California, San Francisco, CA 94110
| | - Karen P. Beckerman
- *Gladstone Institute of Virology and Immunology, San Francisco, CA 94141, and Departments of Microbiology and Immunology and Medicine, University of California, San Francisco, CA 94110
- ‡Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, CA 94110
| | | | - Joseph M. McCune
- *Gladstone Institute of Virology and Immunology, San Francisco, CA 94141, and Departments of Microbiology and Immunology and Medicine, University of California, San Francisco, CA 94110
| |
Collapse
|
29
|
Su. HIV-1 pathogenesis and therapeutic intervention in the SCID-hu Thy/Liv mouse: a model for primary HIV-1 infection in the human thymus. Rev Med Virol 1997; 7:157-166. [PMID: 10398480 PMCID: PMC4435957 DOI: 10.1002/(sici)1099-1654(199709)7:3<157::aid-rmv197>3.0.co;2-r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The SCID-hu Thy/Liv mouse is a model for the analysis of human thymopoiesis. It has been constructed by engrafting fragments of human fetal liver and thymus into the immunodeficient C.B-17 scid/scid (SCID) mouse. The resulting 'Thy/Liv' organ promotes long-term differentiation of human T cells. Given the apparently normal physiology of the SCID-hu Thy/Liv organ, it has been used to explore the pathophysiologic mechanisms of HIV-1 infection in vivo, and to test therapeutic modalities such as anti-HIV-1 drugs and haematopoietic stem cell (HSC)-based gene therapy. In this review, I will summarise what we have learned from the SCID-hu Thy/Liv model, with a focus on recent findings in HIV-1 replication and therapy. Unique HIV-1 determinants have been identified which are required for replication in the Thy/Liv organ but not for replication in PBMC or in T cell lines in vitro. The mechanism of HIV-1 induced thymus depletion is not clear. It is correlated with high levels of HIV-1 replication. Both direct and indirect mechanisms may be involved. In addition to preclinical evaluation of anti- HIV-1 drugs, the SCID-hu Thy/Liv mouse has also been successfully used to test the feasibility of HSC-based gene therapy. A number of improved SCID-hu models have been constructed to meet different requirements. Using these SCID-hu Thy/Liv models, current/future efforts will provide insightful information for understanding pathogenesis and designing therapeutic interventions against HIV-1 infection in humans, especially in paediatric patients. Copyright 1997 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Su
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599- 7295, USA
| |
Collapse
|
30
|
Bonyhadi ML, Moss K, Voytovich A, Auten J, Kalfoglou C, Plavec I, Forestell S, Su L, Böhnlein E, Kaneshima H. RevM10-expressing T cells derived in vivo from transduced human hematopoietic stem-progenitor cells inhibit human immunodeficiency virus replication. J Virol 1997; 71:4707-16. [PMID: 9151864 PMCID: PMC191692 DOI: 10.1128/jvi.71.6.4707-4716.1997] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A key feature of the pathogenesis of human immunodeficiency virus type 1 (HIV-1) infection is the gradual loss of CD4-positive T cells. A number of gene therapy strategies have been designed with the intent of inhibiting HIV replication in mature T cells. As T cells are products of hematolymphoid differentiation, insertion of antiviral genes into hematopoietic stem cells could serve as a vehicle to confer long-term protection in progeny T cells derived from transduced stem cells. One such "cellular immunization" strategy utilizes the gene coding for the HIV-1 rev trans-dominant mutant protein RevM10 which has been demonstrated to inhibit HIV-1 replication in T-cell lines and in primary T cells. In this study, we used a Moloney murine leukemia virus-based retrovirus encoding a bicistronic message coexpressing RevM10 and the murine CD8-alpha' chain (Lyt2). This vector allows rapid selection of transgene-expressing cells as well as quantitation of transgene expression. We demonstrate that RevM10-transduced CD34-enriched hematopoietic progenitor-stem cells (HPSC) isolated from human umbilical cord blood or from granulocyte colony-stimulating factor-mobilized peripheral blood can give rise to mature thymocytes in the SCID-hu thymus/liver mouse model. The phenotypic distribution of HPSC-derived thymocytes is normal, and expression of the transgene can be detected by flow cytometric analysis. Moreover, we demonstrate that RevM10 can inhibit HIV replication in T cells derived from transduced HPSC after expansion in vitro. This is the first demonstration of anti-HIV efficacy in T cells derived from transduced human HPSC.
Collapse
Affiliation(s)
- M L Bonyhadi
- Progenesys Program, SyStemix, Inc., Palo Alto, California 94304, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Bonyhadi ML, Kaneshima H. The SCID-hu mouse: an in vivo model for HIV-1 infection in humans. MOLECULAR MEDICINE TODAY 1997; 3:246-53. [PMID: 9211415 DOI: 10.1016/s1357-4310(97)01046-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The lack of suitable animal models for the in vivo study of HIV-1 infection has prompted investigators to take advantage of the graft-rejection deficit in severe combined immunodeficient (SCID) mice. Two separate approaches have been used to transplant human lymphoid and/or hemolymphoid tissues into SCID mice to generate chimeric animals in which distinct elements of the human immune system could be maintained and studied in vivo. The two models that arose from this work were the SCID-hu mouse and hu-PBL-SCID mouse. The goal of producing these distinct models is to provide an easily manipulable model for the in vivo analysis of HIV-1 infection and its ensuing pathophysiology. Both models support HIV-1 replication and display potential as models for studying antiviral strategies and mechanisms of viral pathogenesis. This review focuses on the SCID-hu mouse.
Collapse
|
32
|
Touraine JL. In utero transplantation of fetal liver stem cells into human fetuses. JOURNAL OF HEMATOTHERAPY 1996; 5:195-9. [PMID: 8723800 DOI: 10.1089/scd.1.1996.5.195] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Based on our experience in the field of fetal liver transplantation (FLT) that we have developed since 1976, we initiated, in 1988, in utero FLT into human fetuses, taking advantage of the immunologic tolerance in young fetuses. The transplants have involved fetuses suffering from various diseases at 12-28 weeks postfertilization, with 2 of the 6 cases eventually resulting in abortion. With the 4 other fetuses, a favorable outcome was observed. Three children are more than 4 years old, and they are alive and well, with evidence of engraftment, reconstitution of immunity, and partial correction of beta-thalassemia. In the fourth case, the fetus is alive and well and birth is expected soon. In utero transplantation of stem cells is a therapy with remarkable advantages: (a) tolerance induction due to immune immaturity of the host, (b) lack of graft-versus-host disease (GVHD) due to immaturity of the donor, (c) ideal isolation of the fetus in the maternal uterus, and (d) optimal environment for donor fetal cell development in the vicinity of host fetal cells and growth factors.
Collapse
Affiliation(s)
- J L Touraine
- Department of Transplantation & Clinical Immunology, Hôpital Edouard Herriot, Lyon, France
| |
Collapse
|
33
|
Ruiz M, Roodman ST, Bouhasin JD, Knutsen AP. T cell differentiation/maturation of CD34+ stem cells from HIV-seropositive hemophiliacs in cultured thymic epithelial fragments. Stem Cells 1996; 14:132-45. [PMID: 8820959 DOI: 10.1002/stem.140132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The clinical manifestations of AIDS are predominantly due to the cellular and humoral immune dysfunction caused by HIV infection, and thymic dysplasia caused by HIV infection probably contributes to the T cell lymphopenia. In the present study, T cell differentiation and/or maturation was assessed when enriched CD34+ stem cells (SCs or SC) purified from bone marrow of HIV-seropositive hemophiliacs were cocultured with allogeneic cultured thymic epithelial fragments (CTEFs). When HIV-seropositive hemophiliacs' enriched CD34+ SC were cocultured with allogeneic CTEFs, acquisition of the T cell phenotypic markers CD7, CD2, CD3, CD4, CD8 and T cell receptor for antigen (TCR) alpha beta was observed from cells harvested from the culture media peaking at approximately 28 days. Origin of the differentiated and matured T cells from the CD34+ SC was confirmed by labeling the SC with 5-(and -6)-(((4-chloromethyl)benzoyl)amino)tetra-methyl-rhodamine (CMTMR), a fluorescent cytoplasmic dye, and detecting fluorescence in the differentiated and matured T cell by flow cytometry. In one experiment, CMTMR labeling was omitted and double positive CD4+CD8+ and triple positive CD3+CD4+CD8+ thymocytes were identified. These studies confirmed that thymocyte differentiation/maturation from SC had occurred. In addition, T cells obtained from the CD34+ SC and CTEF cocultures proliferated to phytohemagglutinin stimulation maximally with stem cell donor antigen-presenting cells (APCs) and also proliferated to pooled B cells in a mixed lymphocyte culture (MLC). Furthermore, the T cells produced were tolerant to thymus donor B cell HLA antigens (p < 0.025); though there was slight MLC reactivity to autologous stem cell donor B cell HLA compared to thymic B cells (p < 0.025). These T cells demonstrated positive self-alloreactivity to stem cell HLA antigens in four of nine persons, though decreased compared to pool B cell alloantigens. Furthermore, in three experiments, responsiveness to stem cell donor B cells subsequently disappeared upon further duration of CD34+ SC-CTEF coculture. These studies suggested that CD34+ SC gave rise to accessory cells populating the thymus that contributed to HLA restriction. To further evaluate this hypothesis, two different donors of CD34+ SC were cultured simultaneously with thymic epithelial fragments and MLC reactivity was then examined toward APC of the stem cell donors. In these experiments, T cells responded to stimulation with HLA antigens of the pool B cells and did not respond to thymus donor B cells. In six of eight experiments, the chimeric SC-CTEF T cells did not respond to stimulation with B cells of either stem cell donor. These studies suggest that HLA restriction and tolerance were induced by cells of the stem cell donor as well as the thymic epithelial cell HLA antigens. In summary, these studies demonstrated that HIV-infected hemophiliac bone marrow-derived nonadherent CD34+ SC were capable of differentiating and/or maturing into T cells when cocultured in a normal allogeneic thymic environment. Furthermore, the T cells derived from derived CD34+ SC were capable of differentiating into T cells when cocultured in a normal allogeneic thymic environment, proliferated maximally with APCs from the stem cell donor and were tolerant of thymic HLA class II antigens, and to a lesser degree to stem cell donor B cell HLA antigens.
Collapse
Affiliation(s)
- M Ruiz
- Pediatric Research Institute, St. Louis University Health Sciences Center, MO 63104, USA
| | | | | | | |
Collapse
|
34
|
Theofilopoulos AN, Baccalà R, González-Quintial R, Kono DH, Balderas RS, Macphee R, Duncan SR, Roglic M. T-cell repertoires in health and disease. Ann N Y Acad Sci 1995; 756:53-65. [PMID: 7645874 DOI: 10.1111/j.1749-6632.1995.tb44481.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
35
|
de Heer C, Schuurman HJ, Houben GF, Pieters RH, Penninks AH, van Loveren H. The SCID-hu mouse as a tool in immunotoxicological risk assessment: effects of 2-acetyl-4(5)-tetrahydroxybutyl-imidazole (THI) and di-n-butyltin dichloride (DBTC) on the human thymus in SCID-hu mice. Toxicology 1995; 100:203-11. [PMID: 7624878 DOI: 10.1016/0300-483x(95)03093-u] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
SCID mice engrafted with human fetal thymus and liver tissue fragments (SCID-hu mice) are currently considered as a new tool in human immunotoxicological risk assessment. Testing of various immunotoxicants exerting thymotoxicity via different intrathymic target cell types is necessary for validation of this model. Therefore, SCID-hu mice were exposed to 2-acetyl-4(5)-(1,2,3,4-tetrahydroxybutyl)-imidazole (THI), the immunotoxic component in the food additive, Caramel Colour III, or the organotin compound, di-n-butyltin dichloride (DBTC). Histopathological examination of the human thymus grafts of SCID-hu mice either exposed to THI or to DBTC showed a reduction in the relative size of the thymus cortex, an effect also described in rodents. These results indicate that the human thymus is a target for the immunotoxic action of both THI and DBTC. In addition, they indicate the promising potential of the SCID-hu mouse model as a tool for human immunotoxicological risk assessment.
Collapse
Affiliation(s)
- C de Heer
- National Institute of Public Health and Environmental Protection, Bilthoven, The Netherlands
| | | | | | | | | | | |
Collapse
|
36
|
Kumamoto T, Inaba M, Toki J, Adachi Y, Imamura H, Ikehara S. Cytotoxic effects of irradiation and deoxyguanosine on fetal thymus. Immunobiology 1995; 192:365-381. [PMID: 7649568 DOI: 10.1016/s0171-2985(11)80176-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Effects of irradiation and deoxyguanosine on the fetal thymus were examined both in vitro and in vivo. Fetal thymi (gestation day 15) of C57BL/6 mice that had been irradiated (0-25 Gy) or treated with various doses of deoxyguanosine (dGuo) were engrafted under the renal capsules of BALB/c nu/nu mice, and the differentiation of T cells was investigated in the engrafted thymi or spleens of these mice. After in vitro treatment of fetal thymi with 1.35 mM dGuo (which was previously reported to be an optimal dose), T cell precursors still remained in some cultures, whereas 1.80 mM dGuo was highly cytotoxic not only to T cell precursors but also to thymic epithelial cells. In contrast, 25 Gy irradiation totally eliminated the T cell precursors from the fetal thymi, though the capacity of epithelial cells to induce T cell differentiation was retained. Although irradiated thymi had the capacity to induce T cell differentiation when assayed in an in vitro organ culture system, long-term observation of thymi engrafted into BALB/c nu/nu mice revealed that, if they had been irradiated (9.5 Gy or 25 Gy), the thymi became scarred by 12 wks after their transplantation. Furthermore, the expression of cell interaction molecules such as ICAM-1 and MHC class II on the thymus stromal cells decreased after irradiation. The interaction molecules decreased 3 wks after 25 Gy irradiation and 7 wks after 9.5 Gy irradiation. The alteration in T cell subsets in the thymus (decreases in both double- and single- positive cells and an increase in double-negative cells) correlated with the decreases in the interaction molecules. This indicates that irradiation (even 9.5 Gy) impairs the T cell-induction capacity of the thymus stromal cells, resulting in an alteration of the T cell subsets followed by a change in the T cell counts in the thymus. Therefore, the long-term effects of irradiation of the thymus should be considered in cases of fetal thymus grafts or total body irradiation before bone marrow transplantation, particularly in the newborn.
Collapse
Affiliation(s)
- T Kumamoto
- Department of Thoracic Surgery, Kansai Medical University, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Péault B, Tirouvanziam R, Sombardier MN, Chen S, Perricaudet M, Gaillard D. Gene transfer to human fetal pulmonary tissue developed in immunodeficient SCID mice. Hum Gene Ther 1994; 5:1131-7. [PMID: 7530495 DOI: 10.1089/hum.1994.5.9-1131] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Human fetal lung rudiments (8-12 weeks of development) undergo considerable growth upon microsurgical ectopic implantation in the xenograft-tolerant SCID mouse, and differentiate into a lung-like tissue that includes: (i) bronchial structures lined with pseudostratified, secretory, ciliated epithelium surrounded by smooth muscle and cartilage rings, (ii) submucosal glands, and (iii) alveolar sacs. Normal expression of the cystic fibrosis transmembrane conductance regulator (CFTR) protein was detected by immunostaining in those grafts, and similar differentiation was observed from either normal or cystic fibrosis (CF) fetal lung rudiments. Upon microinjection into human CF or normal lung grafts in SCID mice, beta-galactosidase-adenovirus gene constructs were efficiently transduced into epithelial and glandular cells. Such an in vivo replica of the human respiratory tissue may be a useful experimental model to study normal and pathologic lung development, and to assay candidate therapeutic gene constructs preclinically.
Collapse
Affiliation(s)
- B Péault
- Institut d'Embryologie Cellulaire et Moléculaire du CNRS, Nogent-sur-Marne, France
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
The severe combined immunodeficient C.B.-17 scid/scid (SCID) mouse has been widely used to study the normal processes of murine lymphoid differentiation. To create an in vivo model of the human hematolymphoid system, this mouse strain has been engrafted with human organ systems (the SCID-hu mouse) or with human peripheral blood mononuclear cells (the hu-PBL-SCID mouse). These mouse models have now been characterized and used to analyze human infectious diseases, hematopoiesis, malignancies and vaccines.
Collapse
Affiliation(s)
- H Kaneshima
- New Enterprise Research Division, SyStemix Inc, Palo Alto, California 94304
| | | | | |
Collapse
|
39
|
Péault B. Human T-cell lineage development in foetal thymus-engrafted SCID mice. RESEARCH IN IMMUNOLOGY 1994; 145:124-8; discussion 155-8. [PMID: 8079044 DOI: 10.1016/s0923-2494(94)80024-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- B Péault
- Institut d'Embryologie cellulaire et moléculaire du CNRS et du Collège de France, Nogent-sur-Marne
| |
Collapse
|
40
|
Spits H, Bárcena A, Hori T, Sánchez MJ, Phillips JH, Galy A. Early events in human intrathymic T-cell development. RESEARCH IN IMMUNOLOGY 1994; 145:128-34; discussion 155-8. [PMID: 8079045 DOI: 10.1016/s0923-2494(94)80025-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- H Spits
- Netherlands Cancer Institute, Amsterdam
| | | | | | | | | | | |
Collapse
|
41
|
Bacchetta R, Bigler M, Touraine JL, Parkman R, Tovo PA, Abrams J, de Waal Malefyt R, de Vries JE, Roncarolo MG. High levels of interleukin 10 production in vivo are associated with tolerance in SCID patients transplanted with HLA mismatched hematopoietic stem cells. J Exp Med 1994; 179:493-502. [PMID: 7905018 PMCID: PMC2191349 DOI: 10.1084/jem.179.2.493] [Citation(s) in RCA: 319] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Transplantation of HLA mismatched hematopoietic stem cells in patients with severe combined immunodeficiency (SCID) can result in a selective engraftment of T cells of donor origin with complete immunologic reconstitution and in vivo tolerance. The latter may occur in the absence of clonal deletion of donor T lymphocytes able to recognize the host HLA antigens. The activity of these host-reactive T cells is suppressed in vivo, since no graft-vs. -host disease is observed in these human chimeras. Here it is shown that the CD4+ host-reactive T cell clones isolated from a SCID patient transplanted with fetal liver stem cells produce unusually high quantities of interleukin 10 (IL-10) and very low amounts of IL-2 after antigen-specific stimulation in vitro. The specific proliferative responses of the host-reactive T cell clones were considerably enhanced in the presence of neutralizing concentrations of an anti-IL-10 monoclonal antibody, suggesting that high levels of endogenous IL-10 suppress the activity of these cells. These in vitro data correlate with observations made in vivo. Semi-quantitative polymerase chain reaction analysis carried out on freshly isolated peripheral blood mononuclear cells (PBMC) of the patient indicated that the levels of IL-10 messenger RNA (mRNA) expression were strongly enhanced, whereas IL-2 mRNA expression was much lower than that in PBMC of healthy donors. In vivo IL-10 mRNA expression was not only high in the T cells, but also in the non-T cell fraction, indicating that host cells also contributed to the high levels of IL-10 in vivo. Patient-derived monocytes were found to be major IL-10 producers. Although no circulating IL-10 could be detected, freshly isolated monocytes of the patient showed a reduced expression of class II HLA antigens. However, their capacity to stimulate T cells of normal donors in primary mixed lymphocyte cultures was within the normal range. Interestingly, similar high in vivo IL-10 mRNA expressions in the T and non-T cell compartment were also observed in three SCID patients transplanted with fetal liver stem cells and in four SCID patients transplanted with T cell-depleted haploidentical bone marrow stem cells. Taken together, these data indicate that high endogenous IL-10 production is a general phenomenon in SCID patients in whom allogenic stem cell transplantation results in immunologic reconstitution and induction of tolerance. Both donor T cells and host accessory cells contribute to these high levels of IL-10, which would suppress the activity of host-reactive T cell in vivo.
Collapse
Affiliation(s)
- R Bacchetta
- Human Immunology Department, DNAX Research Institute, Palo Alto, California 94303-1104
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Touraine JL, Roncarolo MG, Plotnicky H, Bachetta R, Spits H. T lymphocytes from human chimeras do recognize antigen in the context of allogeneic determinants of the major histocompatibility complex. Immunol Lett 1993; 39:9-12. [PMID: 7511565 DOI: 10.1016/0165-2478(93)90157-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Human stem cells from the fetal liver can be transplanted to immunodeficient patients and reconstitute their immunity by giving rise to immunocompetent T lymphocytes of donor origin. Despite full HLA mismatch between donor and host, the helper T cells and the cytotoxic T cells which develop in these chimeric patients are totally functional. They recognize the antigenic peptides presented in the context of the foreign HLA molecules of the recipient, indicating that donor stem cells have been positively selected in the host environment, probably the thymic epithelial cells. By contrast, negative selection appears to be imposed upon T cells by donor hemopoietic cells, probably macrophages or dendritic cells, migrating from the transplant to the host thymus. Clonal deletion is then responsible for tolerance to donor HLA antigens, while clonal anergy explains tolerance to host HLA antigens.
Collapse
Affiliation(s)
- J L Touraine
- Transplantation and Clinical Immunology Unit, INSERM U80, Hôpital Edouard Herriot, Lyon, France
| | | | | | | | | |
Collapse
|
43
|
Boyd RL, Tucek CL, Godfrey DI, Izon DJ, Wilson TJ, Davidson NJ, Bean AG, Ladyman HM, Ritter MA, Hugo P. The thymic microenvironment. IMMUNOLOGY TODAY 1993; 14:445-59. [PMID: 8216723 DOI: 10.1016/0167-5699(93)90248-j] [Citation(s) in RCA: 342] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- R L Boyd
- Dept of Pathology and Immunology, Monash Medical School, Prahran, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kraft DL, Weissman IL, Waller EK. Differentiation of CD3-4-8- human fetal thymocytes in vivo: characterization of a CD3-4+8- intermediate. J Exp Med 1993; 178:265-77. [PMID: 8315382 PMCID: PMC2191096 DOI: 10.1084/jem.178.1.265] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Human thymocyte differentiation was examined by injecting fetal thymic progenitor populations into human thymic xenografts in SCID-hu mice. Thymic progenitors were fluorescently labeled with the lipophilic dye PKH2. The phenotypes of their progeny could be identified by flow cytometric analysis of cells with a very high fluorescent PKH2 signal. Intrathymic injection of purified triple negative (TN) CD3-4-8- thymocytes resulted in the sequential appearance of CD3-4+8-, CD3-4+8+, and CD3+4+8+ cells, with the subsequent appearance of small numbers of phenotypically mature CD3+4+8- and CD3+4-8+ cells over a 4-d period. Sorted CD3-4+8- thymocytes injected intrathymically rapidly differentiated to CD4+8+ cells. CD4+8+ fetal thymocytes in cell cycle differentiated into phenotypically mature CD3+4+8- and CD3+4-8+ populations, whereas nondividing CD4+8+ cells failed to differentiate after intrathymic transfer. The number of cell divisions that occurred between the injection of TN thymocytes and their progeny at different time points was estimated based on the decrease in the intensity of the PKH2 label. The average length of the cell cycle for the TN population was calculated to be 24 h. The SCID-hu model thus provides a useful tool for studying the kinetics of cell division and differentiation of human thymocytes in vivo.
Collapse
Affiliation(s)
- D L Kraft
- Cancer Biology Research Laboratory, Stanford University School of Medicine, California 94305
| | | | | |
Collapse
|
45
|
Baccala R, Vandekerckhove BA, Jones D, Kono DH, Roncarolo MG, Theofilopoulos AN. Bacterial superantigens mediate T cell deletions in the mouse severe combined immunodeficiency-human liver/thymus model. J Exp Med 1993; 177:1481-5. [PMID: 8478618 PMCID: PMC2190996 DOI: 10.1084/jem.177.5.1481] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The ability to analyze T cell receptor (TCR) thymic repertoire shaping in humans by self and foreign ligands is hampered by the lack of suitable models. We recently documented that the mouse severe combined immunodeficiency (SCID)-human fetal liver/thymus model recapitulates the TCR V beta gene repertoire of human thymocytes. Here, we show that an exogenous superantigen, staphylococcal enterotoxin B, administered to such mice induces clonal deletions in both CD4+8- and CD8+4- cells involving the same human V beta clones that are selected in vitro by this toxin. This model, therefore, may allow comprehensive studies into the effects of microbial and other agents on human T cell thymic selection processes in a biologically relevant setting.
Collapse
Affiliation(s)
- R Baccala
- Department of Immunology, Scripps Research Institute, La Jolla, California 92037
| | | | | | | | | | | |
Collapse
|
46
|
Bacchetta R, Vandekerckhove BA, Touraine JL, Bigler M, Martino S, Gebuhrer L, de Vries JE, Spits H, Roncarolo MG. Chimerism and tolerance to host and donor in severe combined immunodeficiencies transplanted with fetal liver stem cells. J Clin Invest 1993; 91:1067-78. [PMID: 8450037 PMCID: PMC288061 DOI: 10.1172/jci116264] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
We have studied the peripheral T cell repertoire of two patients with severe combined immunodeficiency who were successfully treated with human histocompatibility leukocyte antigen (HLA)-mismatched fetal liver stem cell transplantation. The patients presented a split chimerism. T cells were of donor origin, whereas the B cells/monocytes were of the host phenotype. Interestingly, the natural killer (NK) cells in one patient were donor derived and in the other patient of host origin. The NK cells were functional but did not have antihost or donor reactivity. Despite the HLA mismatch between donor and host cells, complete tolerance was achieved in vivo, and a specific unresponsiveness of peripheral blood mononuclear cells from both patients toward the host cells was demonstrated in vitro. Nevertheless, we could isolate T cell receptor (TCR)alpha beta, CD4+ or CD8+, T cell clones specifically reacting with HLA class I and II molecules of the host. The CD4+ host-reactive T cell clones from both patients produced interleukins 2 and 5, interferon-gamma, granulocyte/macrophage colony-stimulating factor but are specifically defective in interleukin 4 production. The frequencies of CD8+ host-reactive T cells were high, and were in the same range as those observed for CD8+ alloreactive T cells. In contrast, no donor-reactive CD8+ T cells or host or donor-reactive TCR gamma delta + T cells were detected. These data indicate that, after fetal stem cell transplantation, donor-reactive, but not host-reactive cells, are deleted from the T cell repertoire. Therefore, a peripheral mechanism of suppression or clonal anergy, rather than clonal deletion, is involved in maintaining in vivo tolerance toward the host.
Collapse
Affiliation(s)
- R Bacchetta
- Human Immunology Department, DNAX Research Institute, Palo Alto, California 94304
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Vandekerckhove BA, Baccala R, Jones D, Kono DH, Theofilopoulos AN, Roncarolo MG. Thymic selection of the human T cell receptor V beta repertoire in SCID-hu mice. J Exp Med 1992; 176:1619-24. [PMID: 1460421 PMCID: PMC2119440 DOI: 10.1084/jem.176.6.1619] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Implantation of pieces of human fetal liver and thymus into SCID mice results in the development of a human thymus-like organ, in which sustained lymphopoiesis is reproducibly observed. In this model, T cell development can be experimentally manipulated. To study the influence of thymic selection on the development of the human T cell repertoire, the T cell receptor (TCR) V beta gene repertoire of double-positive (CD4+CD8+) and single-positive (CD4+CD8- and CD4-CD8+) T cells was analyzed in the SCID-hu thymus using a multiprobe ribonuclease protection assay. TCR diversity in double-positive SCID-hu thymocytes was found to be comparable with that present in the thymus of the fetal liver donor, did not change with time, and was independent of the origin of the thymus donor. Thymic selection in SCID-hu thymus induces changes in V beta usage by the single-positive CD4+ or CD8+ T cells comparable with those previously reported for single-positive cells present in a normal human thymus. Finally, significant differences were observed in the V beta usage by CD4 or CD8 single-positive T cells that matured from genetically identical stem cells in different thymic environments. Collectively, these data suggest: first, that the generation of TCR diversity at the double-positive stage is determined by the genotype of the stem cells; and second, that polymorphic determinants expressed by thymic epithelium measurably influence the V beta repertoire of mature single-positive T cells.
Collapse
Affiliation(s)
- B A Vandekerckhove
- Department of Human Immunology, DNAX Research Institute, Palo Alto, California 94304
| | | | | | | | | | | |
Collapse
|
48
|
Duchosal MA. SCID mice in the study of human autoimmune diseases. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 1992; 14:159-77. [PMID: 1475742 DOI: 10.1007/bf00195292] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- M A Duchosal
- Department of Immunology-IMM2, Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
49
|
Barry TS, Haynes BF. In vivo models of human lymphopoiesis and autoimmunity in severe combined immune deficient mice. J Clin Immunol 1992; 12:311-24. [PMID: 1358912 DOI: 10.1007/bf00920788] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The discovery of the SCID mouse mutation has been an important advance for the study of human lymphopoiesis and autoimmunity. Further work in the SCID mouse models described in this review should yield important new information related to transplantation of human hematopoietic stem cells across HLA barriers, characterization of hematopoietic development in vivo, and identification of pathogenic human T cell clones in organ-specific autoimmune diseases. If pluripotent hematopoietic stem cells and pathogenic autoimmune T cells can be defined using SCID mouse recipients, this would pave the way for development of novel strategies for bone marrow transplantation and for interventional immunotherapy of autoimmune diseases targeted at the T cell receptor (99).
Collapse
Affiliation(s)
- T S Barry
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710
| | | |
Collapse
|
50
|
Abstract
The expression of MHC class I molecules protects cells against lysis by natural killer (NK) cells. It is possible that NK cells are 'educated' to recognize self MHC class I molecules and that the combination of self peptide and MHC class I molecule blocks NK-mediated lysis. Here, Rogier Versteeg compares and contrasts models of education and self-nonself discrimination by T cells and NK cells, and presents a hypothesis for the evolution of T cells from NK cells.
Collapse
Affiliation(s)
- R Versteeg
- Dept of Human Genetics, University of Amsterdam, The Netherlands
| |
Collapse
|